1
|
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, the NIH Reversing Immune Dysfunction for HIV-1 Eradication (RID-HIV) Collaboratory Group. 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
|
2
|
Crowell TA, Ritz J, Zheng L, Naqvi A, Cyktor JC, Puleo J, Clagett B, Lama JR, Kanyama C, Little SJ, Cohn SE, Riddler SA, Collier AC, Heath SL, Tantivitayakul P, Grinsztejn B, Arduino RC, Rooney JF, van Zyl GU, Coombs RW, Fox L, Ananworanich J, Eron JJ, Sieg SF, Mellors JW, Daar ES, for the AIDS Clinical Trials Group (ACTG) A5354/EARLIER Study Team.. Impact of antiretroviral therapy during acute or early HIV infection on virologic and immunologic outcomes: results from a multinational clinical trial. AIDS 2024; 38:1141-1152. [PMID: 38489580 PMCID: PMC11323228 DOI: 10.1097/qad.0000000000003881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
OBJECTIVE To assess how antiretroviral therapy (ART) initiation during acute or early HIV infection (AEHI) affects the viral reservoir and host immune responses. DESIGN Single-arm trial of ART initiation during AEHI at 30 sites in the Americas, Africa, and Asia. METHODS HIV DNA was measured at week 48 of ART in 5 million CD4 + T cells by sensitive qPCR assays targeting HIV gag and pol . Peripheral blood mononuclear cells were stimulated with potential HIV T cell epitope peptide pools consisting of env , gag , nef, and pol peptides and stained for expression of CD3, CD4, CD8, and intracellular cytokines/chemokines. RESULTS From 2017 to 2019, 188 participants initiated ART during Fiebig stages I ( n = 6), II ( n = 43), III ( n = 56), IV ( n = 23), and V ( n = 60). Median age was 27 years (interquartile range 23-38), 27 (14%) participants were female, and 180 (97%) cisgender. Among 154 virally suppressed participants at week 48, 100% had detectable HIV gag or pol DNA. Participants treated during Fiebig I had the lowest HIV DNA levels ( P < 0.001). Week 48 HIV DNA mostly did not correlate with concurrent CD4 + or CD8 + T cell HIV-specific immune responses (rho range -0.11 to +0.19, all P > 0.025). At week 48, the magnitude, but not polyfunctionality, of HIV-specific T cell responses was moderately reduced among participants who initiated ART earliest. CONCLUSION Earlier ART initiation during AEHI reduced but did not eliminate the persistence of HIV-infected cells in blood. These findings explain the rapid viral rebound observed after ART cessation in early-treated individuals with undetectable HIV DNA by less sensitive methods.
Collapse
Affiliation(s)
- Trevor A. Crowell
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Justin Ritz
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Zheng
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Asma Naqvi
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Joseph Puleo
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | | | | | | | - Susan E. Cohn
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | | | | | | - Roberto C. Arduino
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | | | | | - Lawrence Fox
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jintanat Ananworanich
- Amsterdam UMC, Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - Joseph J. Eron
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Eric S. Daar
- Lundquist Institute at Harbor–UCLA Medical Center, Torrance, CA, USA
| | | |
Collapse
|
3
|
Joy J, Gervassi A, Chen L, Kirshenbaum B, Styrchak S, Ko D, McLaughlin S, Shao D, Kosmider E, Edlefsen PT, Maenza J, Collier AC, Mullins JI, Horton H, Frenkel LM. Antigen specificities and proviral integration sites differ in HIV-infected cells by timing of antiretroviral treatment initiation. J Clin Invest 2024; 134:e159569. [PMID: 38833307 PMCID: PMC11245156 DOI: 10.1172/jci159569] [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: 07/08/2022] [Accepted: 05/24/2024] [Indexed: 06/06/2024] Open
Abstract
Despite effective antiretroviral therapy (ART), persons living with HIV harbor reservoirs of persistently infected CD4+ cells, which constitute a barrier to cure. Initiation of ART during acute infection reduces the size of the HIV reservoir, and we hypothesized that in addition, it would favor integration of proviruses in HIV-specific CD4+ T cells, while initiation of ART during chronic HIV infection would favor relatively more proviruses in herpesvirus-specific cells. We further hypothesized that proviruses in acute ART initiators would be integrated into antiviral genes, whereas integration sites (ISs) in chronic ART initiators would favor genes associated with cell proliferation and exhaustion. We found that the HIV DNA distribution across HIV-specific versus herpesvirus-specific CD4+ T cells was as hypothesized. HIV ISs in acute ART initiators were significantly enriched in gene sets controlling lipid metabolism and HIF-1α-mediated hypoxia, both metabolic pathways active in early HIV infection. Persistence of these infected cells during prolonged ART suggests a survival advantage. ISs in chronic ART initiators were enriched in a gene set controlling EZH2 histone methylation, and methylation has been associated with diminished long terminal repeat transcription. These differences that we found in antigen specificities and IS distributions within HIV-infected cells might be leveraged in designing cure strategies tailored to the timing of ART initiation.
Collapse
Affiliation(s)
- Jaimy Joy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Ana Gervassi
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Lennie Chen
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | | | - Sheila Styrchak
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Daisy Ko
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Sherry McLaughlin
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Danica Shao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ewelina Kosmider
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Paul T. Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | - James I. Mullins
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Department of Medicine
- Department of Global Health
| | - Helen Horton
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Lisa M. Frenkel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Medicine
- Department of Global Health
- Department of Pediatrics, and
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
Mchantaf G, Cheret A, Melard A, Essat A, Gardiennet E, Bauer R, Charre C, Meiffredy V, Piroth L, Goujard C, Meyer L, Avettand-Fenoel V. The build-up of stock of stable integrated proviruses overtime explains the difficulty in reducing HIV-1 DNA levels when treatment is initiated at the chronic stage of the infection. J Virus Erad 2023; 9:100357. [PMID: 38188640 PMCID: PMC10770741 DOI: 10.1016/j.jve.2023.100357] [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: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
Background Understanding factors affecting the size and the evolution of the HIV reservoir is essential for the development of curative strategies. This study aimed to assess the impact of antiretroviral therapy (ART) initiated during primary infection (PHI) vs chronic infection (CHI) on the levels and dynamics of integrated HIV-1 DNA, a biomarker of viral persistence. Methods Integrated and total HIV-1-DNA were measured in the blood of 92 patients treated during PHI (early group) and 41 during CHI (deferred group), at diagnosis, ART initiation, and 12-24 months on treatment. Results On ART, detectable (>1.78 log10 copies/106 PBMCs) integrated HIV-1 DNA levels were significantly lower in the early vs deferred group (2.99 log10vs 3.29 log10,p = 0.005). The proportion of undetectable integrated HIV-1 DNA tended to be higher in the early group vs deferred group (61 % vs 46 %; p = 0.133). Conclusion Treatment initiated at PHI limits the levels of integrated HIV-1 DNA in blood. However, initiating treatment at CHI does not allow reaching such low levels in most patients, probably because the stable proviruses at that stage are present in the less prone to elimination long-lived cells. Thus, early ART could provide an opportunity to preparing for functional cure and eradication strategies.
Collapse
Affiliation(s)
- Gilbert Mchantaf
- CHU d’Orléans, Orléans, France
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
- Université d’Orléans, Orléans, France
| | - Antoine Cheret
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
- Plateforme de Médecine Ambulatoire, CHU Guadeloupe, France
| | - Adeline Melard
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
| | - Asma Essat
- Centre de Recherche en Epidémiologie et Santé des Populations (CESP), INSERM U1018, Université Paris Saclay, Le Kremlin-Bicêtre, France
- AP-HP, Service d’Epidémiologie et de Santé Publique, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Elise Gardiennet
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
| | | | - Caroline Charre
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
- AP-HP, Laboratoire de Virologie, Hôpital Cochin, Paris, France
| | | | - Lionel Piroth
- Infectious Diseases Department, Dijon University Hospital, INSERM CIC 1432, Module Épidémiologie Clinique, Université de Bourgogne, Dijon, France
| | - Cécile Goujard
- Centre de Recherche en Epidémiologie et Santé des Populations (CESP), INSERM U1018, Université Paris Saclay, Le Kremlin-Bicêtre, France
- AP-HP, Service de Médecine Interne et d’Immunologie Clinique, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Laurence Meyer
- Centre de Recherche en Epidémiologie et Santé des Populations (CESP), INSERM U1018, Université Paris Saclay, Le Kremlin-Bicêtre, France
- INSERM SC10-US19, Villejuif, France
- AP-HP, Service d’Epidémiologie et de Santé Publique, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Véronique Avettand-Fenoel
- CHU d’Orléans, Orléans, France
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, Paris, France
- Université d’Orléans, Orléans, France
| |
Collapse
|
5
|
Zhang Q, Jiao YM, Li G, Zhang LX, Gao L, Guo XY, Mijiti Z, Guo YT, Xu W, Huang HH, Wang FS. Serum globulin levels are associated with HIV reservoir size and immune restoration during long-term ART. HIV Med 2023; 24:344-353. [PMID: 36101972 DOI: 10.1111/hiv.13393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 08/08/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The objectives of this study were to analyze the relationship between serum globulin levels and immune restoration and HIV reservoir size during long-term antiretroviral therapy (ART). METHODS We enrolled 13 patients living with HIV who had been receiving ART for 5 years. We measured levels of serum globulin, cell-associated (CA) HIV DNA and RNA, and p24 antibody at 0, 1, 3, and 5 years of ART. CD38 and human leukocyte antigen - DR isotype (HLA-DR) were used as activation markers for T-cell activation. Serum concentrations of the inflammatory cytokines interferon gamma-inducible protein (IP)-10 and soluble CD163 (sCD163) were detected by enzyme-linked immunosorbent assay. We analyzed the relationship between serum globulin levels, HIV reservoir size, immune restoration, T-cell immune activation, and inflammatory levels during long-term ART. RESULTS Our data showed that serum globulin levels in people living with HIV were higher than in healthy controls and significantly decreased during the first year of ART. Serum globulin levels during long-term ART were positively correlated with CA HIV DNA, CA HIV RNA, p24 antibody levels, and CD8+ T-cell counts and negatively correlated with CD4+ T-cell counts and CD4/CD8 ratios. Moreover, serum globulin levels were positively correlated with CD4+ and CD8+ T-cell activation and the concentrations of inflammatory biomarkers IP-10 and sCD163 during long-term ART. CONCLUSIONS Our findings suggest that serum globulin levels may be associated with HIV reservoir size and immune restoration during long-term ART.
Collapse
Affiliation(s)
- Qing Zhang
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.,Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Yan-Mei Jiao
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Guang Li
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lu-Xue Zhang
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.,Infectious Disease Department, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lin Gao
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.,Department of Microbiology & Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing, China
| | - Xiao-Yan Guo
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.,Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zilaiguli Mijiti
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.,Department of Microbiology, School of Basic Medical Sciences, Xinjiang Medical University, Xinjiang, China
| | - Yun-Tian Guo
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wen Xu
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Hui-Huang Huang
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.,Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| |
Collapse
|
6
|
Chu C, Armenia D, Walworth C, Santoro MM, Shafer RW. Genotypic Resistance Testing of HIV-1 DNA in Peripheral Blood Mononuclear Cells. Clin Microbiol Rev 2022; 35:e0005222. [PMID: 36102816 PMCID: PMC9769561 DOI: 10.1128/cmr.00052-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
HIV-1 DNA exists in nonintegrated linear and circular episomal forms and as integrated proviruses. In patients with plasma viremia, most peripheral blood mononuclear cell (PBMC) HIV-1 DNA consists of recently produced nonintegrated virus DNA while in patients with prolonged virological suppression (VS) on antiretroviral therapy (ART), most PBMC HIV-1 DNA consists of proviral DNA produced months to years earlier. Drug-resistance mutations (DRMs) in PBMCs are more likely to coexist with ancestral wild-type virus populations than they are in plasma, explaining why next-generation sequencing is particularly useful for the detection of PBMC-associated DRMs. In patients with ongoing high levels of active virus replication, the DRMs detected in PBMCs and in plasma are usually highly concordant. However, in patients with lower levels of virus replication, it may take several months for plasma virus DRMs to reach detectable levels in PBMCs. This time lag explains why, in patients with VS, PBMC genotypic resistance testing (GRT) is less sensitive than historical plasma virus GRT, if previous episodes of virological failure and emergent DRMs were either not prolonged or not associated with high levels of plasma viremia. Despite the increasing use of PBMC GRT in patients with VS, few studies have examined the predictive value of DRMs on the response to a simplified ART regimen. In this review, we summarize what is known about PBMC HIV-1 DNA dynamics, particularly in patients with suppressed plasma viremia, the methods used for PBMC HIV-1 GRT, and the scenarios in which PBMC GRT has been used clinically.
Collapse
Affiliation(s)
- Carolyn Chu
- Department of Family and Community Medicine, University of California San Francisco, San Francisco, California, USA
| | - Daniele Armenia
- UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy
| | - Charles Walworth
- LabCorp-Monogram Biosciences, South San Francisco, California, USA
| | - Maria M. Santoro
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Robert W. Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
7
|
Kumar G, Cottalorda-Dufayard J, Garraffo R, De Salvador-Guillouët F, Cua E, Roger PM. Raltegravir Inclusion Decreases CD4 T-Cells Intra-Cellular Viral Load and Increases CD4 and CD28 Positive T-Cells in Selected HIV Patients. Cells 2022; 11:cells11020208. [PMID: 35053324 PMCID: PMC8773801 DOI: 10.3390/cells11020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/04/2023] Open
Abstract
Raltegravir (RLT) prevents the integration of HIV DNA in the nucleus, but published studies remain controversial, suggesting that it does not decrease proviral DNA. However, there are only a few studies focused on virus-targeted cells. We aimed our study on the impact of RLT inclusion on total intra-cellular viral DNA (TID) in cellular subsets and immune effects in patients with newly acquired undetectable plasmatic viral load (UVL). Six patients having UVL using an antiretroviral combination for 6 months and CD4 T-cells > 350/mL and <500/mL were selected to receive RLT for 3 months from M0 to M3. Patients had 7 sequential viro-immunological determinations from M-1 to M5. Immune phenotypes were determined by flow cytometry and TID quantification was performed using PCR assay on purified cells. TID (median values) at the initiation of RLT in CD4 T-cells was 117 copies/millions of cells, decreased to 27.5 on M3, and remained thereafter permanently under the cut-off (<10 copies/millions of cells) in 4 out of 6 patients. This was associated with an increase of CD4 and CD4 + CD28+ T-cells and a decrease of HLA-DR expression and apoptosis of CD4 T-cells. RLT inclusion led to decreases in the viral load along with positive immune reconstitution, mainly for CD4 T-cells in HIV patients.
Collapse
Affiliation(s)
- Gaurav Kumar
- Unité 576, Centre Hospitalier Universitaire de Nice, Institut National de la Sante et de la Recherche Medicale, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Correspondence: ; Tel.: +1-(405)-271-2907; Fax: +1-(405)-271-4110
| | - Jacqueline Cottalorda-Dufayard
- Virologie, Hopital l’Archet 2, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
| | - Rodolphe Garraffo
- Pharmacologie, Hopital Pasteur, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
| | - Francine De Salvador-Guillouët
- Infectiologie, Hopital l’Archet 1, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France; (F.D.S.-G.); (E.C.)
| | - Eric Cua
- Infectiologie, Hopital l’Archet 1, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France; (F.D.S.-G.); (E.C.)
| | - Pierre-Marie Roger
- Unité 576, Centre Hospitalier Universitaire de Nice, Institut National de la Sante et de la Recherche Medicale, Universite de Nice-Sophia-Antipolis, 06200 Nice, France;
- Infectiologie, Hopital l’Archet 1, Centre Hospitalier Universitaire de Nice, Universite de Nice-Sophia-Antipolis, 06200 Nice, France; (F.D.S.-G.); (E.C.)
- Service Des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Pointe-à-Pitre, 97159 Pointe-à-Pitre, France
| |
Collapse
|
8
|
Bayón-Gil Á, Puertas MC, Urrea V, Bailón L, Morón-López S, Cobarsí P, Brander C, Mothe B, Martinez-Picado J. HIV-1 DNA decay dynamics in early treated individuals: practical considerations for clinical trial design. J Antimicrob Chemother 2021; 75:2258-2263. [PMID: 32335675 PMCID: PMC7366202 DOI: 10.1093/jac/dkaa139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 01/28/2023] Open
Abstract
Background Initiation of combination antiretroviral therapy (cART) soon after HIV-1 infection limits the establishment of viral reservoirs. Thus, early treated individuals are preferred candidates to evaluate novel viral remission strategies. However, their cART-dependent HIV-1 DNA decay dynamics are still poorly defined. This can hamper the design and interpretation of results from clinical trials intended to further reduce viral reservoirs. Objectives To clarify the duration of cART needed for the HIV-1 reservoir to be stabilized in early treated individuals. Methods We characterized the longitudinal decline of total HIV-1 DNA levels by droplet digital PCR in 21 individuals initiating cART within 6 months after estimated HIV-1 acquisition. Measurements were taken at cART initiation, after 6 months and annually until Year 4. Correlations between virological and clinical parameters were statistically analysed. Statistical modelling was performed applying a mixed-effects model. Results Total HIV-1 DNA experienced a median overall decrease of 1.43 log10 units (IQR = 1.17–1.69) throughout the 4 years of follow-up. Baseline levels for total HIV-1 DNA, viral load, absolute CD4+ T cell count and CD4+/CD8+ ratio correlate with final HIV-1 DNA measurements (R2 = 0.68, P < 0.001; R2 = 0.54, P = 0.012; R2 = −0.47, P = 0.031; and R2 = −0.59, P = 0.0046, respectively). Statistical modelling shows that after 2 years on cART the viral reservoir had reached a set point. Conclusions A waiting period of 2 years on cART should be considered when designing interventions aiming to impact latent HIV-1 reservoir levels and viral rebound kinetics after cART discontinuation, in order to facilitate interpretation of results and enhance the chance of viral control.
Collapse
Affiliation(s)
| | | | - Víctor Urrea
- IrsiCaixa AIDS Research Institute, Badalona, Spain
| | - Lucía Bailón
- Fight AIDS Foundation (FLS), Infectious Disease Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | | | - Patricia Cobarsí
- Fight AIDS Foundation (FLS), Infectious Disease Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.,Aelix Therapeutics, Barcelona, Spain
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,Fight AIDS Foundation (FLS), Infectious Disease Service, Germans Trias i Pujol University Hospital, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
9
|
Mak G, Zaunders JJ, Bailey M, Seddiki N, Rogers G, Leong L, Phan TG, Kelleher AD, Koelsch KK, Boyd MA, Danta M. Preservation of Gastrointestinal Mucosal Barrier Function and Microbiome in Patients With Controlled HIV Infection. Front Immunol 2021; 12:688886. [PMID: 34135912 PMCID: PMC8203413 DOI: 10.3389/fimmu.2021.688886] [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: 03/31/2021] [Accepted: 05/04/2021] [Indexed: 01/02/2023] Open
Abstract
Background Despite successful ART in people living with HIV infection (PLHIV) they experience increased morbidity and mortality compared with HIV-negative controls. A dominant paradigm is that gut-associated lymphatic tissue (GALT) destruction at the time of primary HIV infection leads to loss of gut integrity, pathological microbial translocation across the compromised gastrointestinal barrier and, consequently, systemic inflammation. We aimed to identify and measure specific changes in the gastrointestinal barrier that might allow bacterial translocation, and their persistence despite initiation of antiretroviral therapy (ART). Method We conducted a cross-sectional study of the gastrointestinal (GIT) barrier in PLHIV and HIV-uninfected controls (HUC). The GIT barrier was assessed as follows: in vivo mucosal imaging using confocal endomicroscopy (CEM); the immunophenotype of GIT and circulating lymphocytes; the gut microbiome; and plasma inflammation markers Tumour Necrosis Factor-α (TNF-α) and Interleukin-6 (IL-6); and the microbial translocation marker sCD14. Results A cohort of PLHIV who initiated ART early, during primary HIV infection (PHI), n=5), and late (chronic HIV infection (CHI), n=7) infection were evaluated for the differential effects of the stage of ART initiation on the GIT barrier compared with HUC (n=6). We observed a significant decrease in the CD4 T-cell count of CHI patients in the left colon (p=0.03) and a trend to a decrease in the terminal ileum (p=0.13). We did not find evidence of increased epithelial permeability by CEM. No significant differences were found in microbial translocation or inflammatory markers in plasma. In gut biopsies, CD8 T-cells, including resident intraepithelial CD103+ cells, did not show any significant elevation of activation in PLHIV, compared to HUC. The majority of residual circulating activated CD38+HLA-DR+ CD8 T-cells did not exhibit gut-homing integrins α4ß7, suggesting that they did not originate in GALT. A significant reduction in the evenness of species distribution in the microbiome of CHI subjects (p=0.016) was observed, with significantly higher relative abundance of the genus Spirochaeta in PHI subjects (p=0.042). Conclusion These data suggest that substantial, non-specific increases in epithelial permeability may not be the most important mechanism of HIV-associated immune activation in well-controlled HIV-positive patients on antiretroviral therapy. Changes in gut microbiota warrant further study.
Collapse
Affiliation(s)
- Gerald Mak
- St. Vincent's Clinical School, UNSW, Darlinghurst, NSW, Australia
| | - John J Zaunders
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| | | | - Nabila Seddiki
- IDMIT Department/IBFJ, Immunology of Viral Infections and Autoimmune Diseases (IMVA), INSERM U1184, CEA, Université Paris Sud, Paris, France
| | - Geraint Rogers
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.,Faculty of Science, Flinders University, Adelaide, SA, Australia
| | - Lex Leong
- Microbiology and Infectious Diseases, South Australia (SA) Pathology, Adelaide, SA, Australia
| | - Tri Giang Phan
- St. Vincent's Clinical School, UNSW, Darlinghurst, NSW, Australia.,Immunology Division Garvan Institute of Medical Research, Sydney, NSW, Australia
| | | | | | - Mark A Boyd
- Kirby Institute, UNSW Sydney, Sydney, NSW, Australia.,South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.,Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Mark Danta
- St. Vincent's Clinical School, UNSW, Darlinghurst, NSW, Australia.,Department of Gastroenterology, St. Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
10
|
Massanella M, Bender Ignacio RA, Lama JR, Pagliuzza A, Dasgupta S, Alfaro R, Rios J, Ganoza C, Pinto-Santini D, Gilada T, Duerr A, Chomont N. Long-term effects of early antiretroviral initiation on HIV reservoir markers: a longitudinal analysis of the MERLIN clinical study. THE LANCET. MICROBE 2021; 2:e198-e209. [PMID: 35544209 PMCID: PMC8622834 DOI: 10.1016/s2666-5247(21)00010-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/09/2020] [Accepted: 01/14/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Early antiretroviral therapy (ART) initiation (ie, within 3 months of infection) limits establishment of the HIV reservoir. However, the effect of early ART initiation on the long-term dynamics of the pool of infected cells remains unclear. METHODS In this longitudinal analysis, we included cisgender men who have sex with men (MSM) and transgender women (aged 18-54 years) at high risk for HIV infection, enrolled in the ongoing longitudinal MERLIN study in Peru between Oct 28, 2014, and Nov 8, 2018. Participants were eligible if they had been infected with HIV less than 90 days before enrolment, and if they had cryopreserved peripheral blood mononuclear cell (PBMC) samples. Participants were stratified into three groups on the basis of whether they initiated ART at 30 days or less (acute group), at 31-90 days (early group), or more than 24 weeks (deferred group) after the estimated date of detectable infection. PBMC samples were collected before ART initiation and longitudinally for up to 4 years on ART. The main outcomes were to establish the size of the HIV reservoir before ART initiation and to assess the effect of the timing of ART initiation on the decay of the HIV reservoir over 4 years follow-up. We quantified viral load, and isolated CD4 cells to quantify total HIV DNA, integrated HIV DNA and 2-long terminal repeat circles. Longitudinal analysis of active and inducible HIV reservoirs were measured by quantifying the frequency of CD4 cells producing multiply-spliced HIV RNA ex vivo and after in-vitro stimulation with a tat/rev induced limiting dilution assay (TILDA). A mixed-effects model from the time of ART initiation was used to measure longitudinal decays in viral loads and each HIV reservoir measure in each of the three groups. FINDINGS We included 56 participants in this analysis, all of whom were MSM: 15 were in the acute group, 19 were in the early group, and 22 were in the deferred group. Participants in all three groups had similar levels of all HIV reservoir markers before ART initiation. All participants, including those in the acute group, had a pool of transcriptionally silent HIV-infected cells before ART initiation, as indicated by a substantial increase in TILDA measures upon stimulation. Longitudinal analysis over 4 years of ART revealed a biphasic decay of all HIV persistence markers, with a rapid initial decline followed by a slower decay in all participants. During the first-phase decay, the half-lives of both total and integrated HIV DNA and TILDA measures were significantly shorter in the acute group than in the early and deferred groups. During the second-phase decay, HIV reservoir markers continued to decline only in participants in the acute group. INTERPRETATION Participants who initiated ART within 30 days or less of HIV infection showed a steeper and more sustained decay in HIV reservoir measures, suggesting long-term benefit of acute ART initiation on reservoir clearance. FUNDING The US National Institutes of Health and the Canadian Institutes for Health Research.
Collapse
Affiliation(s)
- Marta Massanella
- Centre de Recherche du CHUM, Montreal, QC, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | | | - Javier R Lama
- Asociación Civil Impacta Salud y Educación, Lima, Perú
| | - Amélie Pagliuzza
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Sayan Dasgupta
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Jessica Rios
- Asociación Civil Impacta Salud y Educación, Lima, Perú
| | | | | | - Trupti Gilada
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ann Duerr
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; University of Washington, Seattle, WA, USA
| | - Nicolas Chomont
- Centre de Recherche du CHUM, Montreal, QC, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
11
|
CD73 + CD127 high Long-Term Memory CD4 T Cells Are Highly Proliferative in Response to Recall Antigens and Are Early Targets in HIV-1 Infection. Int J Mol Sci 2021; 22:ijms22020912. [PMID: 33477692 PMCID: PMC7831934 DOI: 10.3390/ijms22020912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
HIV-1 infection rapidly leads to a loss of the proliferative response of memory CD4+ T lymphocytes, when cultured with recall antigens. We report here that CD73 expression defines a subset of resting memory CD4+ T cells in peripheral blood, which highly express the α-chain of the IL-7 receptor (CD127), but not CD38 or Ki-67, yet are highly proliferative in response to mitogen and recall antigens, and to IL-7, in vitro. These cells also preferentially express CCR5 and produce IL-2. We reasoned that CD73+ memory CD4+ T cells decrease very early in HIV-1 infection. Indeed, CD73+ memory CD4+ T cells comprised a median of 7.5% (interquartile range: 4.5-10.4%) of CD4+ T cells in peripheral blood from healthy adults, but were decreased in primary HIV-1 infection to a median of 3.7% (IQR: 2.6-6.4%; p = 0.002); and in chronic HIV-1 infection to 1.9% (IQR: 1.1-3%; p < 0.0001), and were not restored by antiretroviral therapy. Moreover, we found that a significant proportion of CD73+ memory CD4+ T cells were skewed to a gut-homing phenotype, expressing integrins α4 and β7, CXCR3, CCR6, CD161 and CD26. Accordingly, 20% of CD4+ T cells present in gut biopsies were CD73+. In HIV+ subjects, purified CD73+ resting memory CD4+ T cells in PBMC were infected with HIV-1 DNA, determined by real-time PCR, to the same level as for purified CD73-negative CD4+ T cells, both in untreated and treated subjects. Therefore, the proliferative CD73+ subset of memory CD4+ T cells is disproportionately reduced in HIV-1 infection, but, unexpectedly, their IL-7 dependent long-term resting phenotype suggests that residual infected cells in this subset may contribute significantly to the very long-lived HIV proviral DNA reservoir in treated subjects.
Collapse
|
12
|
Shelton EM, Reeves DB, Bender Ignacio RA. Initiation of Antiretroviral Therapy during Primary HIV Infection: Effects on the Latent HIV Reservoir, Including on Analytic Treatment Interruptions. AIDS Rev 2020; 23:28-39. [PMID: 33105471 PMCID: PMC7987773 DOI: 10.24875/aidsrev.20000001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022]
Abstract
Antiretroviral therapy (ART) inhibits HIV replication but does not eradicate the latent reservoir. The previous research suggests that earlier ART initiation provides benefit on limiting reservoir size, but timing and extent of this effect remain unclear. Analytic treatment interruption (ATI) may be used to demonstrate HIV remission, but whether early ART also improves likelihood or duration of even temporary virologic remission is unclear. This review seeks to answer both questions. We performed a systematic review and analysis following Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines and included 21 interventional or observational studies with sufficient HIV reservoir outcomes. We also aggregated reservoir outcomes and transformed data into approximate measurements of total HIV DNA per million peripheral blood mononuclear cells and analyzed the correlation between timing of ART initiation and reservoir size. People living with HIV who initiate ART in primary infection maintain smaller reservoirs on suppressive ART than those who initiate treatment during chronic infection. The reduction of reservoir is most pronounced when ART is started within 2 weeks of HIV acquisition. Across studies, we found a moderately strong association between longer time to ART initiation and reservoir size, which was strongest when measured after 1 year on ART (Pearson's r = 0.69, p = 0.0003). After ATI, larger pre-ATI reservoir size predicts shorter time to viral rebound. Early ART may also facilitate long-term control of viremia. Although achieving sustained HIV remission will require further interventions, initiating ART very early in infection could limit the extent of the reservoir and also lead to post-ATI control in rare cases.
Collapse
Affiliation(s)
- Eva M. Shelton
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Daniel B. Reeves
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Rachel A. Bender Ignacio
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, USA
| |
Collapse
|
13
|
Mapping the extent of heterogeneity of human CCR5+ CD4+ T cells in peripheral blood and lymph nodes. AIDS 2020; 34:833-848. [PMID: 32044843 DOI: 10.1097/qad.0000000000002503] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND CD4 T cells that express the chemokine receptor, CCR5, are the most important target of HIV-1 infection, but their functions, phenotypes and anatomical locations are poorly understood. We aimed to use multiparameter flow cytometry to better define the full breadth of these cells. METHODS High-parameter fluorescence flow and mass cytometry were optimized to analyse subsets of CCR5 memory CD4 T cells, including CD25CD127 Tregs, CXCR3CCR6- Th1-like, CCR6CD161CXCR3- Th17-like, integrins α4ß7 gut-homing, CCR4 skin-homing, CD62L lymph node-homing, CD38HLA-DR activated cells, and CD27-CD28- cytotoxic T lymphocytes, in a total of 22 samples of peripheral blood, ultrasound-guided fine needle biopsies of lymph nodes and excised tonsils. CCR5 antigen-specific CD4 T cells were studied using the OX40 flow-based assay. RESULTS 10-20% of CCR5 memory CD4 T cells were Tregs, 10-30% were gut-homing, 10-30% were skin-homing, 20-40% were lymph node-homing, 20-50% were Th1-like and 20-40% were Th17-like cells. Up to 30% were cytotoxic T lymphocytes in CMV-seropositive donors, including cells that were either CCR5Granzyme K or CCR5Granzyme B. When all possible phenotypes were exhaustively analysed, more than 150 different functional and trafficking subsets of CCR5 CD4 T cells were seen. Moreover, a small population of resident CD69Granzyme KCCR5 CD4 T cells was found in lymphoid tissues. CMV- and Mycobacterium tuberculosis-specific CD4 T cells were predominantly CCR5. CONCLUSION These results reveal for the first time the prodigious heterogeneity of function and trafficking of CCR5 CD4 T cells in blood and in lymphoid tissue, with significant implications for rational approaches to prophylaxis for HIV-1 infection and for purging of the HIV-1 reservoir in those participants already infected.
Collapse
|
14
|
Delagreverie HM, Bauduin C, De Castro N, Grinsztejn B, Chevrier M, Jouenne F, Mourah S, Kalidi I, Pilotto JH, Brites C, Tregnago Barcellos N, Amara A, Wittkop L, Molina JM, Delaugerre C. Impact of Raltegravir or Efavirenz on Cell-Associated Human Immunodeficiency Virus-1 (HIV-1) Deoxyribonucleic Acid and Systemic Inflammation in HIV-1/Tuberculosis Coinfected Adults Initiating Antiretroviral Therapy. Open Forum Infect Dis 2020; 7:ofz549. [PMID: 32083147 PMCID: PMC7019658 DOI: 10.1093/ofid/ofz549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/02/2020] [Indexed: 01/01/2023] Open
Abstract
Background In view of the fast viremia decline obtained with integrase inhibitors, we studied the respective effects of initiating efavirenz (EFV) or raltegravir (RAL)-based antiretroviral therapy (ART) regimens on human immunodeficiency virus (HIV)-1 deoxyribonucleic acid (DNA) levels and inflammation biomarkers in the highly inflammatory setting of advanced HIV-1 disease with tuberculosis (TB) coinfection. Methods We followed cell-associated HIV-1 DNA, high-sensitivity C-reactive protein (hsCRP), interleukin 6 (IL-6), soluble CD14 and D-Dimer levels for 48 weeks after ART initiation in the participants to the ANRS12-180 REFLATE-TB study. This phase II open-label randomized study included ART-naive people with HIV and TB treated with rifampicin to receive RAL 400 mg twice daily (RAL400), RAL 800 mg twice daily (RAL800) or EFV 600 mg QD with tenofovir and lamivudine. Results In 146 participants, the median (interquartile range [IQR]) week (W)0 HIV-1 DNA level was 4.7 (IQR, 4.3–5.1) log10 copies/106 CD4+, and the reduction by W48 was −0.8 log10 copies/106 CD4+ on EFV, −0.9 on RAL400, and −1.0 on RAL800 (P = .74). Baseline median (IQR) hsCRP, IL-6, sCD14, and D-Dimer levels were 6.9 (IQR, 3.3–15.6) mg/L, 7.3 (IQR, 3.5–12.3) pg/mL, 3221 (IQR, 2383–4130) ng/mL, and 975 (IQR, 535–1970) ng/mL. All biomarker levels decreased over the study: the overall W0–W48 mean (95% confidence interval) fold-change on ART was 0.37 (IQR, 0.28–0.48) for hsCRP, 0.42 (IQR, 0.35–0.51) for IL-6, 0.51 (IQR, 0.47–0.56) for sCD14, and 0.39 (IQR, 0.32–0.47) for D-Dimers. There were no differences in biomarker reduction across treatment arms. Conclusions In participants with HIV and TB, EFV, RAL400, or RAL800 effectively and equally reduced inflammation and HIV-1 DNA levels.
Collapse
Affiliation(s)
- Héloïse M Delagreverie
- Laboratoire de Virologie, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France.,INSERM U944, Université de Paris, Paris, France
| | - Claire Bauduin
- ISPED, Inserm, Bordeaux Population Health Research Center, Team MORPH3EUS, UMR 1219, CIC-EC 1401, Bordeaux University, Bordeaux, France
| | - Nathalie De Castro
- Maladies Infectieuses et Tropicales, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Beatriz Grinsztejn
- Evandro Chagas Clinical Research Institute-Fiocruz, STD/AIDS Clinical Research Laboratory, Rio de Janeiro, Brazil
| | - Marc Chevrier
- Laboratoire de Biochimie, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Fanélie Jouenne
- Laboratoire de Pharmacologie, Hôpital Saint-Louis Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Samia Mourah
- Laboratoire de Pharmacologie, Hôpital Saint-Louis Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Issa Kalidi
- Laboratoire d'Hématologie, Hôpital Saint-Louis Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Carlos Brites
- Hospital Universitário Profesor Edgar Santos, Laboratório de Pesquisa em Doenças Infecciosas, Bahia, Brazil
| | | | - Ali Amara
- INSERM U944, Université de Paris, Paris, France
| | - Linda Wittkop
- ISPED, Inserm, Bordeaux Population Health Research Center, Team MORPH3EUS, UMR 1219, CIC-EC 1401, Bordeaux University, Bordeaux, France.,Pole de Santé Publique, Service d'Information Medicale, Bordeaux, France
| | - Jean-Michel Molina
- INSERM U944, Université de Paris, Paris, France.,Maladies Infectieuses et Tropicales, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Constance Delaugerre
- Laboratoire de Virologie, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France.,INSERM U944, Université de Paris, Paris, France
| |
Collapse
|
15
|
Bon I, Calza L, Musumeci G, Longo S, Bertoldi A, D'Urbano V, Gibellini D, Magistrelli E, Viale PL, Re MC. Impact of Different Antiretroviral Strategies on Total HIV-DNA Level in Virologically Suppressed HIV-1 Infected Patients. Curr HIV Res 2019; 15:448-455. [PMID: 29210661 PMCID: PMC5876918 DOI: 10.2174/1570162x16666171206121026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/02/2017] [Accepted: 11/28/2017] [Indexed: 01/11/2023]
Abstract
Background: Total HIV-DNA load in peripheral blood cell (PBMCs) reflects the global viral reservoir that seems not to be affected by antiretroviral treatment. However, some studies report-ed a different permeability of different drugs in cellular compartments. Objective: To investigate the relation between the amount of total HIV-1 DNA and different treatment strategies. Methods: Total HIV-1 DNA was quantified by real time PCR in PBMCs collected from 161 patients with long-term undetectable HIV-RNA receiving different therapy schedules (3-drug regimens or 2-drug regimen containing Raltegravir as integrase inhibitor). Results: Overall, HIV patients who started therapy with a median pre-ART CD4+ cell count >400 cells/mm3 and HIV viral load of 3 log10 copies/ml, achieved a lower amount of HIV total DNA. No significant correlation was found in DNA size when patients were stratified on the basis of different therapeutic protocols. However, HIV DNA load analysis, when only performed in HIV patients with a median pre-ART CD4+ cell count >200 cells/mm3 and HIV viral load < 3 log10 copies/ml, showed a significative DNA decrease in Raltegravir treated group with respect to the NNRTIs-treated group. Conclusion: The data emphasize that HIV-DNA level represents a predictive factor in long-term sup-pressive therapy patients. In addition, the diminished reservoir, only observed in patients treated with the NRTI-sparing regimen RAL plus PI/r before immunological and virological derangement, sug-gests that latest generation drugs, such as integrase inhibitors, might represent an optimal chance in the management of HIV infection.
Collapse
Affiliation(s)
- Isabella Bon
- Microbiology Section of the Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Leonardo Calza
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Giuseppina Musumeci
- Microbiology Section of the Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Serena Longo
- Microbiology Section of the Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Alessia Bertoldi
- Microbiology Section of the Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Vanessa D'Urbano
- Microbiology Section of the Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Davide Gibellini
- Unit of Microbiology Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Eleonora Magistrelli
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Pier Luigi Viale
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy
| | - Maria Carla Re
- Microbiology Section of the Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, University of Bologna, Via Massarenti, 9, Bologna, Italy.,Interuniversity Consortium, National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| |
Collapse
|
16
|
Zaunders J, Dyer WB, Churchill M, Munier CML, Cunningham PH, Suzuki K, McBride K, Hey-Nguyen W, Koelsch K, Wang B, Hiener B, Palmer S, Gorry PR, Bailey M, Xu Y, Danta M, Seddiki N, Cooper DA, Saksena NK, Sullivan JS, Riminton S, Learmont J, Kelleher AD. Possible clearance of transfusion-acquired nef/LTR-deleted attenuated HIV-1 infection by an elite controller with CCR5 Δ32 heterozygous and HLA-B57 genotype. J Virus Erad 2019. [DOI: 10.1016/s2055-6640(20)30056-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
17
|
Luo L, Wang N, Yue Y, Han Y, Lv W, Liu Z, Qiu Z, Lu H, Tang X, Zhang T, Zhao M, He Y, Shenghua H, Wang M, Li Y, Huang S, Li Y, Liu J, Tuofu Z, Routy JP, Li T. The effects of antiretroviral therapy initiation time on HIV reservoir size in Chinese chronically HIV infected patients: a prospective, multi-site cohort study. BMC Infect Dis 2019; 19:257. [PMID: 30871484 PMCID: PMC6419375 DOI: 10.1186/s12879-019-3847-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/22/2019] [Indexed: 12/31/2022] Open
Abstract
Background The effect of ART initiation time on HIV-1 DNA reservoir in chronically infected individuals is not well understood. Determining the potential influencing factors associated with a low HIV-1 DNA level in chronic infection is an important step toward drug-free control. Methods A prospective study included 444 chronically HIV-infected adults was performed. Participants were divided into two groups: early initiation group (EIG) or delayed initiation group (DIG) based on their baseline CD4 count; 350 to 500 and < 350 cells/mm3, respectively. Total HIV-1 DNA was measured by quantitative PCR. Using the Mann-Whitney U test, the HIV-1 DNA level at week 48 was compared between the two groups. The influencing factors of the HIV-1 DNA and factors associated with achieving a low HIV-1 level at week 48 were analyzed. Results The HIV-1 DNA at week 48 in EIG was significantly lower than in DIG [2.12 (1.80–2.51) vs 2.58 (2.21–2.87) log10 copies/106peripheral blood mononuclear cells (PBMCs); p = 0.001]. Early ART initiation was positively associated with lower HIV-1 DNA at week 48 (p = 0.025). Similarly, baseline HIV-1DNA (p = 0.001) was positively associated with HIV-1DNA at week 48 and baseline CD4/CD8 ratio (p = 0.001) was inversely associated with HIV-1DNA at week 48. Early ART initiation (p = 0.003) and baseline HIV-1 DNA level (p < 0.001) were positively associated with achieving HIV-1 DNA < 100 copies/106 PBMCs at week 48. Conclusion Early ART initiation is positively associated with a smaller size of viral reservoir and a higher possibility of achieving a low HIV-1DNA level at week 48 in Chinese chronically HIV-1 infected adult. Trial registration NCT01844297; Registered 1 May, 2013.
Collapse
Affiliation(s)
- Ling Luo
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Nidan Wang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Yongsong Yue
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Yang Han
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Wei Lv
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Zhengyin Liu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Zhifeng Qiu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Hongzhou Lu
- Shanghai Public Health Clinical Center affiliated with Fudan University, Shanghai, China
| | | | - Tong Zhang
- Beijing You'an Hospital, Capital Medical University, Beijing, China
| | - Min Zhao
- 302 Military Hospital of China, Beijing, China
| | - Yun He
- The Infectious Disease Hospital of Henan Province, Zhengzhou, China
| | - He Shenghua
- Chengdu Infectious Diseases Hospital, Chengdu, China
| | - Min Wang
- The First Hospital of Changsha, Changsha, China
| | - Yongzhen Li
- The Center for Disease Prevention and Control of Guangxi province, Nanning, China
| | | | - Yong Li
- The Longtan Hospital, Liuzhou, China
| | - Jing Liu
- The hospital affiliated with the Chinese Medical University, Hangzhou, China
| | - Zhu Tuofu
- Department of Laboratory Medicine, University of Washington, Seattle, USA
| | - Jean-Pierre Routy
- Research Institute of the McGill University Health Centre, Chronic Viral Illness Service, and Division of Hematology, McGill University Health Centre, Montreal, Québec, Canada
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| |
Collapse
|
18
|
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
|
19
|
Hey-Nguyen WJ, Bailey M, Xu Y, Suzuki K, Van Bockel D, Finlayson R, Leigh Brown A, Carr A, Cooper DA, Kelleher AD, Koelsch KK, Zaunders JJ. HIV-1 DNA Is Maintained in Antigen-Specific CD4+ T Cell Subsets in Patients on Long-Term Antiretroviral Therapy Regardless of Recurrent Antigen Exposure. AIDS Res Hum Retroviruses 2019; 35:112-120. [PMID: 30511878 DOI: 10.1089/aid.2018.0235] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Memory CD4+ T cells (mCD4s) containing integrated HIV DNA are considered the main barrier to a cure for HIV infection. Here, we analyzed HIV DNA reservoirs in antigen-specific subsets of mCDs to delineate the mechanisms by which HIV reservoirs persist during antiretroviral therapy (ART). HIV Gag, cytomegalovirus (CMV), and tetanus toxoid (TT)-specific mCD4s were isolated from peripheral blood samples obtained from 11 individual subjects, 2-11 years after commencing ART. Antigen-specific mCD4s were identified by the sensitive OX40 assay and purified by cell sorting. Total HIV DNA levels were quantified by real-time PCR, and clonal viral sequences generated from mCD4 subsets and pre-ART plasma samples. Quantitative results and sequence analysis were restricted to five and three study participants, respectively, which was likely due to the low frequency of the antigen-specific mCD4s and relatively low HIV DNA proviral loads. Median HIV Gag-, CMV-, and TT-specific mCD4s were 0.61%, 2.46%, and 0.78% of total mCD4s, and they contained a median of 2.50, 2.38, and 2.55 log10 copies of HIV DNA per 106 cells, respectively. HIV DNA sequences were derived from antigen-specific mCD4s clustered with sequences derived from pre-ART plasma samples. There was a trend toward increased viral diversity in clonal viral sequences derived from CMV-specific mCD4s relative to TT-specific mCD4s. Despite limitations, this study provides direct evidence that HIV reservoirs persist in memory CD4+ T cell subsets maintained by homeostatic proliferation (TT) and adds to growing evidence against viral evolution during ART. Similar future studies require techniques that sample diverse HIV reservoirs and with improved sensitivity.
Collapse
Affiliation(s)
- William J. Hey-Nguyen
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
| | - Michelle Bailey
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
| | - Yin Xu
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
| | - Kazuo Suzuki
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
- St. Vincent's Hospital Sydney, Sydney, Australia
| | - David Van Bockel
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
- St. Vincent's Hospital Sydney, Sydney, Australia
| | - Robert Finlayson
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Taylor Square Private Clinic, Sydney, Australia
| | - Andrew Leigh Brown
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew Carr
- St. Vincent's Hospital Sydney, Sydney, Australia
| | - David A. Cooper
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
| | - Anthony D. Kelleher
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
- St. Vincent's Hospital Sydney, Sydney, Australia
| | - Kersten K. Koelsch
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
- St. Vincent's Hospital Sydney, Sydney, Australia
| | - John J. Zaunders
- The Kirby Institute, UNSW Australia, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital Sydney, Sydney, Australia
- St. Vincent's Hospital Sydney, Sydney, Australia
| |
Collapse
|
20
|
|
21
|
Méndez C, Ledger S, Petoumenos K, Ahlenstiel C, Kelleher AD. RNA-induced epigenetic silencing inhibits HIV-1 reactivation from latency. Retrovirology 2018; 15:67. [PMID: 30286764 PMCID: PMC6172763 DOI: 10.1186/s12977-018-0451-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/01/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Current antiretroviral therapy is effective in controlling HIV-1 infection. However, cessation of therapy is associated with rapid return of viremia from the viral reservoir. Eradicating the HIV-1 reservoir has proven difficult with the limited success of latency reactivation strategies and reflects the complexity of HIV-1 latency. Consequently, there is a growing need for alternate strategies. Here we explore a "block and lock" approach for enforcing latency to render the provirus unable to restart transcription despite exposure to reactivation stimuli. Reactivation of transcription from latent HIV-1 proviruses can be epigenetically blocked using promoter-targeted shRNAs to prevent productive infection. We aimed to determine if independent and combined expression of shRNAs, PromA and 143, induce a repressive epigenetic profile that is sufficiently stable to protect latently infected cells from HIV-1 reactivation when treated with a range of latency reversing agents (LRAs). RESULTS J-Lat 9.2 cells, a model of HIV-1 latency, expressing shRNAs PromA, 143, PromA/143 or controls were treated with LRAs to evaluate protection from HIV-1 reactivation as determined by levels of GFP expression. Cells expressing shRNA PromA, 143, or both, showed robust resistance to viral reactivation by: TNF, SAHA, SAHA/TNF, Bryostatin/TNF, DZNep, and Chaetocin. Given the physiological importance of TNF, HIV-1 reactivation was induced by TNF (5 ng/mL) and ChIP assays were performed to detect changes in expression of epigenetic markers within chromatin in both sorted GFP- and GFP+ cell populations, harboring latent or reactivated proviruses, respectively. Ordinary two-way ANOVA analysis used to identify interactions between shRNAs and chromatin marks associated with repressive or active chromatin in the integrated provirus revealed significant changes in the levels of H3K27me3, AGO1 and HDAC1 in the LTR, which correlated with the extent of reduced proviral reactivation. The cell line co-expressing shPromA and sh143 consistently showed the least reactivation and greatest enrichment of chromatin compaction indicators. CONCLUSION The active maintenance of epigenetic silencing by shRNAs acting on the HIV-1 LTR impedes HIV-1 reactivation from latency. Our "block and lock" approach constitutes a novel way of enforcing HIV-1 "super latency" through a closed chromatin architecture that renders the virus resistant to a range of latency reversing agents.
Collapse
Affiliation(s)
- Catalina Méndez
- Department of Immunovirology and Pathogenesis, Level 5, Wallace Wurth Building, The Kirby Institute for Infection and Immunity, UNSW Sydney, Kensington, Sydney, NSW, 2052, Australia
| | - Scott Ledger
- Department of Immunovirology and Pathogenesis, Level 5, Wallace Wurth Building, The Kirby Institute for Infection and Immunity, UNSW Sydney, Kensington, Sydney, NSW, 2052, Australia
| | - Kathy Petoumenos
- Department of Immunovirology and Pathogenesis, Level 5, Wallace Wurth Building, The Kirby Institute for Infection and Immunity, UNSW Sydney, Kensington, Sydney, NSW, 2052, Australia
| | - Chantelle Ahlenstiel
- Department of Immunovirology and Pathogenesis, Level 5, Wallace Wurth Building, The Kirby Institute for Infection and Immunity, UNSW Sydney, Kensington, Sydney, NSW, 2052, Australia.
| | - Anthony D Kelleher
- Department of Immunovirology and Pathogenesis, Level 5, Wallace Wurth Building, The Kirby Institute for Infection and Immunity, UNSW Sydney, Kensington, Sydney, NSW, 2052, Australia
| |
Collapse
|
22
|
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
|
23
|
Murray JM, Zaunders J, Emery S, Cooper DA, Hey-Nguyen WJ, Koelsch KK, Kelleher AD. HIV dynamics linked to memory CD4+ T cell homeostasis. PLoS One 2017; 12:e0186101. [PMID: 29049331 PMCID: PMC5648138 DOI: 10.1371/journal.pone.0186101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/25/2017] [Indexed: 11/18/2022] Open
Abstract
The dynamics of latent HIV is linked to infection and clearance of resting memory CD4+ T cells. Infection also resides within activated, non-dividing memory cells and can be impacted by antigen-driven and homeostatic proliferation despite suppressive antiretroviral therapy (ART). We investigated whether plasma viral level (pVL) and HIV DNA dynamics could be explained by HIV’s impact on memory CD4+ T cell homeostasis. Median total, 2-LTR and integrated HIV DNA levels per μL of peripheral blood, for 8 primary (PHI) and 8 chronic HIV infected (CHI) individuals enrolled on a raltegravir (RAL) based regimen, exhibited greatest changes over the 1st year of ART. Dynamics slowed over the following 2 years so that total HIV DNA levels were equivalent to reported values for individuals after 10 years of ART. The mathematical model reproduced the multiphasic dynamics of pVL, and levels of total, 2-LTR and integrated HIV DNA in both PHI and CHI over 3 years of ART. Under these simulations, residual viremia originated from reactivated latently infected cells where most of these cells arose from clonal expansion within the resting phenotype. Since virion production from clonally expanded cells will not be affected by antiretroviral drugs, simulations of ART intensification had little impact on pVL. HIV DNA decay over the first year of ART followed the loss of activated memory cells (120 day half-life) while the 5.9 year half-life of total HIV DNA after this point mirrored the slower decay of resting memory cells. Simulations had difficulty reproducing the fast early HIV DNA dynamics, including 2-LTR levels peaking at week 12, and the later slow loss of total and 2-LTR HIV DNA, suggesting some ongoing infection. In summary, our modelling indicates that much of the dynamical behavior of HIV can be explained by its impact on memory CD4+ T cell homeostasis.
Collapse
Affiliation(s)
- John M. Murray
- School of Mathematics and Statistics, UNSW Australia, Sydney, NSW, Australia
- * E-mail:
| | - John Zaunders
- St Vincent's Hospital, Sydney, Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | - Sean Emery
- The Kirby Institute, University of New South Wales, Sydney, NSW Australia
| | - David A. Cooper
- The Kirby Institute, University of New South Wales, Sydney, NSW Australia
| | | | - Kersten K. Koelsch
- The Kirby Institute, University of New South Wales, Sydney, NSW Australia
| | | |
Collapse
|
24
|
Parisi SG, Andreis S, Basso M, Cavinato S, Scaggiante R, Franzetti M, Andreoni M, Palù G, Cattelan AM. Time course of cellular HIV-DNA and low-level HIV viremia in HIV-HCV co-infected patients whose HCV infection had been successfully treated with directly acting antivirals. Med Microbiol Immunol 2017; 206:419-428. [PMID: 28864951 DOI: 10.1007/s00430-017-0518-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/27/2017] [Indexed: 12/21/2022]
Abstract
This longitudinal study described cellular HIV-DNA changes and their correlation with HIV low-level plasma viremia (LLV) in HIV-HCV co-infected patients on successful antiretroviral and anti-HCV therapy by treatment with direct-acting antivirals (DAA). Thirty-nine patients were examined prior to the start of DAA (T0), after week 12 (T1) and 24 weeks (T2) of anti-HCV therapy. Cellular PBMC HIV-DNA was analysed as an absolute value and as the percentage of increase or decrease from T0 to T2. Patients were classified as having undetectable plasma HIV viraemia (UV) or LLV in the year before the start of anti-HCV treatment and within the T0-T2 study period. Thirty-five patients (89.7%) of the 39 subjects enrolled had the same plasma HIV viraemia control in the year before HCV treatment and in the T0-T2 interval. The HIV-DNA value at T0 and at T2 was higher in patients with LLV than in subjects with UV (p = 0.015 and p = 0.014, respectively). A similar proportion of patients with LLV and UV experienced an increase or decrease of HIV-DNA from T0 to T2. The percentage increase in HIV-DNA value (262.8%) from T0 to T2 was higher compared to the decrease (43.5%) in patients with UV (p = 0.012), and it was higher compared to the percentage increase in HIV-DNA value reported in subjects with LLV (262.8 versus 49%, p = 0.026). HIV-HCV co-infected patients experienced a multifaceted perturbation of cellular HIV-DNA levels within a 24-week period during anti-HCV treatment; the extent of the phenomenon was greater in subjects with UV. Fast HCV-RNA clearance seemed to have a greater influence on the cellular reservoir than on plasma HIV-RNA.
Collapse
Affiliation(s)
- Saverio G Parisi
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35100, Padua, Italy.
| | - Samantha Andreis
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35100, Padua, Italy
| | - Monica Basso
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35100, Padua, Italy
| | - Silvia Cavinato
- Infectious Diseases Unit, Padova Hospital, Via Giustiniani, 2, 35128, Padua, Italy
| | - Renzo Scaggiante
- Infectious Diseases Unit, Padova Hospital, Via Giustiniani, 2, 35128, Padua, Italy
| | - Marzia Franzetti
- Infectious Diseases Unit, Padova Hospital, Via Giustiniani, 2, 35128, Padua, Italy
| | - Massimo Andreoni
- Clinical Infectious Diseases, Tor Vergata University, Viale Oxford, 81, 00133, Rome, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35100, Padua, Italy
| | - Anna Maria Cattelan
- Infectious Diseases Unit, Padova Hospital, Via Giustiniani, 2, 35128, Padua, Italy
| |
Collapse
|
25
|
Sun Y, Fu Y, Zhang Z, Tang T, Liu J, Ding H, Han X, Xu J, Chu Z, Shang H, Jiang Y. The investigation of CD4+T-cell functions in primary HIV infection with antiretroviral therapy. Medicine (Baltimore) 2017; 96:e7430. [PMID: 28700479 PMCID: PMC5515751 DOI: 10.1097/md.0000000000007430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection leads to reduced CD4T-cell counts and immune dysfunction. Initiation of antiretroviral therapy (ART) in HIV primary infection has been recommended to achieve an optimal clinical outcome, but a comprehensive study on restoration of CD4T-cell function in primary HIV-infected individuals with ART still needs to be eluciated. We investigated longitudinal changes in the CD4T-cell counts, phenotypes, and functions in HIV-infected individuals with early ART (initiated within 6 months after HIV infection) or later ART (initiated more than 12 months after HIV infection). Patients from early ART and later ART groups had received ART for at least 1 year. Individuals with early ART had more CD4T cells, a faster rate of CD4T-cell recovery than those receiving later ART; the levels of CD4T-cell activation and senescence were lower in early ART compared to those with later ART (P = .031; P = .016), but the activation was higher than normal controls (NC) (P = .001); thymic emigrant function was more upregulated in early ART than in later ART (P = .015), but still lower than NC (P = .027); proliferative capacity and interferon-γ secretion of CD4T cells were significantly decreased in primary infection (P < .001; P = .029), and early ART restored these CD4T-cell functions, there is no difference with NC, later ART could partially restore the functions of CD4T cells, but it remained lower than that of NC (P = .005; P = .019). Early ART could better improve CD4T-cell function.
Collapse
|
26
|
Hey-Nguyen WJ, Xu Y, Pearson CF, Bailey M, Suzuki K, Tantau R, Obeid S, Milner B, Field A, Carr A, Bloch M, Cooper DA, Kelleher AD, Zaunders JJ, Koelsch KK. Quantification of Residual Germinal Center Activity and HIV-1 DNA and RNA Levels Using Fine Needle Biopsies of Lymph Nodes During Antiretroviral Therapy. AIDS Res Hum Retroviruses 2017; 33:648-657. [PMID: 28287825 DOI: 10.1089/aid.2016.0171] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
HIV-1 reservoirs are most often studied in peripheral blood (PB), but not all lymphocytes recirculate, particularly T follicular helper (Tfh) CD4+ T cells, as well as germinal center (GC) B cells, in lymph nodes (LNs). Ultrasound-guided fine needle biopsies (FNBs) from inguinal LNs and PB samples were obtained from 10 healthy controls (HCs) and 21 HIV-1-infected subjects [11 antiretroviral therapy (ART) naive and 10 on ART]. Tfh cells and GC B cells were enumerated by flow cytometry. HIV-1 DNA and cell-associated (CA) RNA levels in LNs and PB were quantified by real-time polymerase chain reaction. FNBs were obtained without adverse events. Tfh cells and GC B cells were highly elevated in ART-naive subjects, with a median GC B cell count >300-fold higher than HCs, but also remained higher in 4 out of the 10 subjects on ART. GC B cell counts and Tfh cell counts were highly correlated with each other, and also with activated T cells in LNs but not in blood. Levels of HIV-1 DNA and CA RNA viral burden in highly purified CD4+ T cells from FNBs were significantly elevated compared with those in CD4+ T cells from PB in the ART-naive group, but only trended toward an increase in the ART patients. FNBs enabled minimally invasive access to, and parallel measurement of residual activated T and B cells and viral burden within LNs in HIV-1-infected patients. These FNBs revealed significant GC activity that was not apparent from corresponding PB samples.
Collapse
Affiliation(s)
| | - Yin Xu
- Kirby Institute, UNSW Sydney, Sydney, Australia
| | | | | | - Kazuo Suzuki
- Kirby Institute, UNSW Sydney, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital, Darlinghurst, Australia
| | - Robyn Tantau
- Department of Medical Imaging, St. Vincent's Hospital, Darlinghurst, Australia
| | - Solange Obeid
- Department of Medical Imaging, St. Vincent's Hospital, Darlinghurst, Australia
| | - Brad Milner
- Department of Medical Imaging, St. Vincent's Hospital, Darlinghurst, Australia
| | - Andrew Field
- Department of Anatomical Pathology, St. Vincent's Hospital, Darlinghurst, Australia
| | - Andrew Carr
- Centre for Applied Medical Research, St. Vincent's Hospital, Darlinghurst, Australia
| | - Mark Bloch
- Holdsworth House Medical Practice, Darlinghurst, Australia
| | - David A. Cooper
- Kirby Institute, UNSW Sydney, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital, Darlinghurst, Australia
| | - Anthony D. Kelleher
- Kirby Institute, UNSW Sydney, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital, Darlinghurst, Australia
| | - John J. Zaunders
- Kirby Institute, UNSW Sydney, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital, Darlinghurst, Australia
| | - Kersten K. Koelsch
- Kirby Institute, UNSW Sydney, Sydney, Australia
- Centre for Applied Medical Research, St. Vincent's Hospital, Darlinghurst, Australia
| |
Collapse
|
27
|
Total HIV-1 DNA, a Marker of Viral Reservoir Dynamics with Clinical Implications. Clin Microbiol Rev 2017; 29:859-80. [PMID: 27559075 DOI: 10.1128/cmr.00015-16] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
HIV-1 DNA persists in infected cells despite combined antiretroviral therapy (cART), forming viral reservoirs. Recent trials of strategies targeting latent HIV reservoirs have rekindled hopes of curing HIV infection, and reliable markers are thus needed to evaluate viral reservoirs. Total HIV DNA quantification is simple, standardized, sensitive, and reproducible. Total HIV DNA load influences the course of the infection and is therefore clinically relevant. In particular, it is predictive of progression to AIDS and death, independently of HIV RNA load and the CD4 cell count. Baseline total HIV DNA load is predictive of the response to cART. It declines during cART but remains quantifiable, at a level that reflects both the history of infection (HIV RNA zenith, CD4 cell count nadir) and treatment efficacy (residual viremia, cumulative viremia, immune restoration, immune cell activation). Total HIV DNA load in blood is also predictive of the presence and severity of some HIV-1-associated end-organ disorders. It can be useful to guide individual treatment, notably, therapeutic de-escalation. Although it does not distinguish between replication-competent and -defective latent viruses, the total HIV DNA load in blood, tissues, and cells provides insights into HIV pathogenesis, probably because all viral forms participate in host cell activation and HIV pathogenesis. Total HIV DNA is thus a biomarker of HIV reservoirs, which can be defined as all infected cells and tissues containing all forms of HIV persistence that participate in pathogenesis. This participation may occur through the production of new virions, creating new cycles of infection and disseminating infected cells; maintenance or amplification of reservoirs by homeostatic cell proliferation; and viral transcription and synthesis of viral proteins without new virion production. These proteins can induce immune activation, thus participating in the vicious circle of HIV pathogenesis.
Collapse
|
28
|
Aggarwal A, Hitchen TL, Ootes L, McAllery S, Wong A, Nguyen K, McCluskey A, Robinson PJ, Turville SG. HIV infection is influenced by dynamin at 3 independent points in the viral life cycle. Traffic 2017; 18:392-410. [PMID: 28321960 DOI: 10.1111/tra.12481] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 12/19/2022]
Abstract
CD4 T cells are important cellular targets for HIV-1, yet the primary site of HIV fusion remains unresolved. Candidate fusion sites are either the plasma membrane or from within endosomes. One area of investigation compounding the controversy of this field, is the role of the protein dynamin in the HIV life cycle. To understand the role of dynamin in primary CD4 T cells we combined dynamin inhibition with a series of complementary assays based on single particle tracking, HIV fusion, detection of HIV DNA products and active viral transcription. We identify 3 levels of dynamin influence on the HIV life cycle. Firstly, dynamin influences productive infection by preventing cell cycle progression. Secondly, dynamin influences endocytosis rates and increases the probability of endosomal fusion. Finally, we provide evidence in resting CD4 T cells that dynamin directly regulates the HIV fusion reaction at the plasma membrane. We confirm this latter observation using 2 divergent dynamin modulating compounds, one that enhances dynamin conformations associated with dynamin ring formation (ryngo-1-23) and the other that preferentially targets dynamin conformations that appear in helices (dyngo-4a). This in-depth understanding of dynamin's roles in HIV infection clarifies recent controversies and furthermore provides evidence for dynamin regulation specifically in the HIV fusion reaction.
Collapse
Affiliation(s)
- Anupriya Aggarwal
- The Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Tina L Hitchen
- The Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Lars Ootes
- The Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Samantha McAllery
- The Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Andrew Wong
- The Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Khanh Nguyen
- The Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Adam McCluskey
- Centre for Chemical Biology, Chemistry, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, Australia
| | - Phillip J Robinson
- Children's Medical Research Institute, The University of Sydney, New South Wales, Australia
| | - Stuart G Turville
- The Kirby Institute, University of New South Wales, New South Wales, Australia
| |
Collapse
|
29
|
Ruggiero A, Malatinkova E, Rutsaert S, Paxton WA, Vandekerckhove L, De Spiegelaere W. Utility of integrated HIV-1 DNA quantification in cure studies. Future Virol 2017. [DOI: 10.2217/fvl-2016-0130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Numerous HIV-1 curative strategies have been proposed to eradicate the virus reservoir pool that remains integrated within target cells, despite successful antiretroviral therapy. To test the impact of such interventions on this reservoir, a universal marker of persistence is needed. Quantifying integrated HIV-1 DNA load has been proposed as a strong virological marker. In this paper, we provide a detailed description of the most commonly used assays to quantify integrated HIV-1 DNA and applications in relevant clinical studies produced over the last 20 years with a major focus on the recent literature. We discuss the potential for using this marker of virological persistence and the technical limitations that need to be addressed.
Collapse
Affiliation(s)
- Alessandra Ruggiero
- Department of Clinical Infection, Microbiology & Immunology (CIMI), Institute of Infection & Global Health, University of Liverpool, Liverpool, UK
| | - Eva Malatinkova
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine & Health Sciences, Ghent University, Belgium
| | - Sofie Rutsaert
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine & Health Sciences, Ghent University, Belgium
| | - William A Paxton
- Department of Clinical Infection, Microbiology & Immunology (CIMI), Institute of Infection & Global Health, University of Liverpool, Liverpool, UK
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine & Health Sciences, Ghent University, Belgium
| | - Ward De Spiegelaere
- Department of Morphology, Faculty of Veterinary Sciences, Ghent University, Belgium
| |
Collapse
|
30
|
Achieving HIV-1 Control through RNA-Directed Gene Regulation. Genes (Basel) 2016; 7:genes7120119. [PMID: 27941595 PMCID: PMC5192495 DOI: 10.3390/genes7120119] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/28/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
HIV-1 infection has been transformed by combined anti-retroviral therapy (ART), changing a universally fatal infection into a controllable infection. However, major obstacles for an HIV-1 cure exist. The HIV latent reservoir, which exists in resting CD4+ T cells, is not impacted by ART, and can reactivate when ART is interrupted or ceased. Additionally, multi-drug resistance can arise. One alternate approach to conventional HIV-1 drug treatment that is being explored involves gene therapies utilizing RNA-directed gene regulation. Commonly known as RNA interference (RNAi), short interfering RNA (siRNA) induce gene silencing in conserved biological pathways, which require a high degree of sequence specificity. This review will provide an overview of the silencing pathways, the current RNAi technologies being developed for HIV-1 gene therapy, current clinical trials, and the challenges faced in progressing these treatments into clinical trials.
Collapse
|
31
|
Monitoring Integration over Time Supports a Role for Cytotoxic T Lymphocytes and Ongoing Replication as Determinants of Reservoir Size. J Virol 2016; 90:10436-10445. [PMID: 27630237 DOI: 10.1128/jvi.00242-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/15/2016] [Indexed: 01/30/2023] Open
Abstract
The dynamics of HIV reservoir accumulation off antiretroviral therapy (ART) is underexplored. Levels of integrated HIV DNA in peripheral blood mononuclear cells (PBMCs) were longitudinally monitored before and after antiviral therapy. HIV integration increased over time in both elite controllers (ECs; n = 8) and noncontrollers (NCs; n = 6) before ART, whereas integration remained stable in patients on ART (n = 4). The median annual fold change was higher in NCs than in ECs and negatively correlated with CD4/CD8 T-cell ratio. Cytotoxic T lymphocyte (CTL) function as assessed by infected CD4 T-cell elimination (ICE) and granzyme B activity did not significantly change over time in ECs, suggesting that the gradual increase in integrated HIV DNA observed in ECs was not a result of progressive loss of immune-mediated control. Also, acutely infected (n = 7) but not chronically infected (n = 6) patients exhibited a significant drop in integrated HIV DNA 12 months after ART initiation. In conclusion, in the absence of ART, integrated HIV accumulates over time both in NCs and in ECs, at variable individual rates. Starting ART early in infection leads to a greater drop in integrated HIV DNA than does initiating treatment after years of infection. The increase in integrated HIV DNA over time suggests that early treatment may be of benefit in limiting HIV reservoirs. IMPORTANCE The establishment of a latent reservoir represents a barrier to cure among HIV-infected individuals. The dynamics of HIV reservoir accumulation over time in patients before antiviral therapy is underexplored, in large part because it is difficult to accurately and reproducibly measure the size of HIV reservoir in this setting. In our study, we compared the dynamics of integrated HIV DNA over time in ECs and NCs before and after ART was initiated. We found that integrated HIV DNA levels progressively increase over time in the absence of ART, but with a higher, albeit variable, rate in NCs compared to ECs. In addition, integrated HIV DNA declines more dramatically when ART is initiated in acute rather than chronic HIV infection, suggesting important differences between acute and chronic infection. Our study highlights the role of HIV replication and CTL control in reservoir accumulation in sanctuary sites and why ART appears to be more effective in acute infection.
Collapse
|
32
|
Zaunders J, Danta M, Bailey M, Mak G, Marks K, Seddiki N, Xu Y, Templeton DJ, Cooper DA, Boyd MA, Kelleher AD, Koelsch KK. CD4 + T Follicular Helper and IgA + B Cell Numbers in Gut Biopsies from HIV-Infected Subjects on Antiretroviral Therapy Are Similar to HIV-Uninfected Individuals. Front Immunol 2016; 7:438. [PMID: 27822211 PMCID: PMC5075890 DOI: 10.3389/fimmu.2016.00438] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/04/2016] [Indexed: 01/09/2023] Open
Abstract
Background Disruption of gastrointestinal tract epithelial and immune barriers contribute to microbial translocation, systemic inflammation, and progression of HIV-1 infection. Antiretroviral therapy (ART) may lead to reconstitution of CD4+ T cells in gut-associated lymphoid tissue (GALT), but its impact on humoral immunity within GALT is unclear. Therefore, we studied CD4+ subsets, including T follicular helper cells (Tfh), as well as resident B cells that have switched to IgA production, in gut biopsies, from HIV+ subjects on suppressive ART compared to HIV-negative controls (HNC). Methods Twenty-three HIV+ subjects on ART and 22 HNC undergoing colonoscopy were recruited to the study. Single-cell suspensions were prepared from biopsies from left colon (LC), right colon (RC), and terminal ileum (TI). T and B lymphocyte subsets, as well as EpCAM+ epithelial cells, were accurately enumerated by flow cytometry, using counting beads. Results No significant differences in the number of recovered epithelial cells were observed between the two subject groups. However, the median TI CD4+ T cell count/106 epithelial cells was 2.4-fold lower in HIV+ subjects versus HNC (19,679 versus 47,504 cells; p = 0.02). Similarly, median LC CD4+ T cell counts were reduced in HIV+ subjects (8,358 versus 18,577; p = 0.03) but were not reduced in RC. Importantly, we found no significant differences in Tfh or IgA+ B cell counts at either site between HIV+ subjects and HNC. Further analysis showed no difference in CD4+, Tfh, or IgA+ B cell counts between subjects who commenced ART in primary compared to chronic HIV-1 infection. Despite the decrease in total CD4 T cells, we could not identify a selective decrease of other key subsets of CD4+ T cells, including CCR5+ cells, CD127+ long-term memory cells, CD103+ tissue-resident cells, or CD161+ cells (surrogate marker for Th17), but there was a slight increase in the proportion of T regulatory cells. Conclusion While there were lower absolute CD4+ counts in the TI and LC in HIV+ subjects on ART, they were not associated with significantly reduced Tfh cell counts or IgA+ B cells, suggesting that this important vanguard of adaptive immune defense against luminal microbial products is normalized following ART.
Collapse
Affiliation(s)
- John Zaunders
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia; The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Mark Danta
- St Vincent's Hospital, Clinical School , Sydney, NSW , Australia
| | - Michelle Bailey
- The Kirby Institute, The University of New South Wales , Sydney, NSW , Australia
| | - Gerald Mak
- St Vincent's Hospital, Clinical School , Sydney, NSW , Australia
| | - Katherine Marks
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital , Sydney, NSW , Australia
| | - Nabila Seddiki
- Equipe 16, INSERM U955, Créteil, France; Faculté de médecine, Université Paris Est, Créteil, France; Vaccine Research Institute (VRI), Créteil, France
| | - Yin Xu
- The Kirby Institute, The University of New South Wales , Sydney, NSW , Australia
| | - David J Templeton
- The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia; RPA Sexual Health, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - David A Cooper
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia; The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Mark A Boyd
- The Kirby Institute, The University of New South Wales , Sydney, NSW , Australia
| | - Anthony D Kelleher
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia; The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Kersten K Koelsch
- The Kirby Institute, The University of New South Wales , Sydney, NSW , Australia
| |
Collapse
|
33
|
Michelini Z, Galluzzo CM, Pirillo MF, Francisci D, Degli Antoni A, Vivarelli A, Ladisa N, Cirioni O, Weimer LE, Fragola V, Cara A, Floridia M, Baroncelli S. HIV-1 DNA dynamics and variations in HIV-1 DNA protease and reverse transcriptase sequences in multidrug-resistant patients during successful raltegravir-based therapy. J Med Virol 2016; 88:2115-2124. [PMID: 27197719 DOI: 10.1002/jmv.24581] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2016] [Indexed: 01/04/2023]
Abstract
There is limited information on the variations of HIV-1 DNA mutation profile in reverse transcriptase (RT) and protease (PR) genes during suppressive antiretroviral treatment (plasma HIV-1 RNA continuously <50 copies/ml) with raltegravir (RAL)-based regimens in patients with baseline RT/PR resistant HIV. Twelve multidrug resistant (RT: 12/12, PR: 8/12) HIV-infected patients were followed during effectively suppressive RAL-based therapy. Total and integrated HIV-1 DNA were assessed by real time PCR at baseline and every 6 months. Ultrasensitive (threshold: 2.5 copies/ml) plasma HIV-1 RNA and genotypic analysis of RT and PR in proviral DNA were performed at baseline and at 24 months. Half of the patients had full viral suppression (plasma HIV-RNA < 2.5 copies/ml) at month 12. Total HIV-1 DNA declined significantly after 12 months of therapy (from 249.2 to 145.7 copies/106 cells, P = 0.023), and remained stable until 24 months, when total HIV-1 DNA levels raised, concomitantly with a less stringent suppression of HIV-1 RNA (81.8% of patients with >2.5 copies/ml). Integrated HIV-1 DNA did not show fluctuations during the study period. Sequencing of the PR and RT regions from HIV-1 DNA revealed changes in the resistance mutation profile in five patients. Total HIV-1 DNA declined after the introduction of RAL-based therapy, with a rebound after 2 years. No changes were observed in levels of integrated DNA, suggesting limited effect on archived HIV. The RT and PR sequence changes in archived HIV-1 DNA suggest that variation of the mutation profile can occur even in the absence of detectable HIV-1 RNA. J. Med. Virol. 88:2115-2124, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zuleika Michelini
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Clementina Maria Galluzzo
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Franca Pirillo
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Daniela Francisci
- Division of Infectious Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Anna Degli Antoni
- Department of Infectious Diseases and Hepatology, Azienda Ospedaliera di Parma, Parma, Italy
| | | | | | - Oscar Cirioni
- Clinic of Infectious Diseases, Ospedali Riuniti, Marche Polytechnic University, Ancona, Italy
| | - Liliana Elena Weimer
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Vincenzo Fragola
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Floridia
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Baroncelli
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
34
|
Bermejo M, López-Huertas MR, García-Pérez J, Climent N, Descours B, Ambrosioni J, Mateos E, Rodríguez-Mora S, Rus-Bercial L, Benkirane M, Miró JM, Plana M, Alcamí J, Coiras M. Dasatinib inhibits HIV-1 replication through the interference of SAMHD1 phosphorylation in CD4+ T cells. Biochem Pharmacol 2016; 106:30-45. [PMID: 26851491 DOI: 10.1016/j.bcp.2016.02.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/02/2016] [Indexed: 02/07/2023]
Abstract
Massive activation of infected CD4+ T cells during acute HIV-1 infection leads to reservoir seeding and T-cell destruction. During T-cell activation, the antiviral effect of the innate factor SAMHD1 is neutralized through phosphorylation at T592, allowing HIV-1 infection. Dasatinib, a tyrosine kinase inhibitor currently used for treating chronic myeloid leukemia, has been described to control HIV-1 replication through its negative effect on T-cell proliferation and viral entry. We demonstrate that Dasatinib can actually interfere with SAMHD1 phosphorylation in human peripheral blood lymphocytes, preserving its antiviral activity against HIV-1. Dasatinib prevented SAMHD1 phosphorylation in vitro and ex vivo, impairing HIV-1 reverse transcription and proviral integration. This was the major mechanism of action because the presence of Vpx, which degrades SAMHD1, in HIV-1 virions impeded the inhibitory effect of Dasatinib on HIV-1 replication. In fact, infection with VSV-pseudotyped HIV-1 virions and fusion of BlaM-Vpr-containing HIV-1 viruses with activated PBMCs in the presence of Dasatinib suggested that Dasatinib was not acting at fusion level. Finally, PBMCs from patients on chronic treatment with Dasatinib showed a lower level of SAMHD1 phosphorylation in response to activating stimuli and low susceptibility to HIV-1 infection ex vivo. Consequently, Dasatinib is a compound currently used in clinic that preserves the antiviral function of SAMHD1. Using Dasatinib as adjuvant of antiretroviral therapy during early primary HIV-1 infection would contribute to reduce viral replication and spread, prevent reservoir seeding, and preserve CD4 counts and CTL responses. These events would create a more favorable virologic and immunologic environment for future interventional studies aiming at HIV-1 eradication.
Collapse
MESH Headings
- Adult
- Anti-HIV Agents/pharmacology
- Antineoplastic Agents/pharmacology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/enzymology
- CD4-Positive T-Lymphocytes/virology
- Cell Proliferation/drug effects
- Dasatinib/pharmacology
- Female
- Gene Expression Regulation
- HIV Infections/drug therapy
- HIV Infections/enzymology
- HIV Infections/genetics
- HIV Infections/virology
- HIV-1/drug effects
- HIV-1/genetics
- HIV-1/growth & development
- Host-Pathogen Interactions
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Lymphocyte Activation
- Male
- Monomeric GTP-Binding Proteins/antagonists & inhibitors
- Monomeric GTP-Binding Proteins/genetics
- Monomeric GTP-Binding Proteins/metabolism
- Phosphorylation/drug effects
- Protein Kinase Inhibitors/pharmacology
- SAM Domain and HD Domain-Containing Protein 1
- Signal Transduction
- Vesiculovirus/genetics
- Viral Fusion Proteins/genetics
- Viral Fusion Proteins/metabolism
- Virus Internalization/drug effects
- Virus Replication/drug effects
Collapse
Affiliation(s)
- Mercedes Bermejo
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - María Rosa López-Huertas
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier García-Pérez
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Núria Climent
- Retrovirology and Viral Immunopathology Laboratory, AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Benjamin Descours
- Laboratory of Molecular Virology, Institute of Human Genetics, Montpellier, France
| | - Juan Ambrosioni
- Infectious Diseases Service, AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Elena Mateos
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Rodríguez-Mora
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Lucía Rus-Bercial
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Monsef Benkirane
- Laboratory of Molecular Virology, Institute of Human Genetics, Montpellier, France
| | - José M Miró
- Infectious Diseases Service, AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Montserrat Plana
- Retrovirology and Viral Immunopathology Laboratory, AIDS Research Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - José Alcamí
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Mayte Coiras
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
35
|
Sarmati L, D'Ettorre G, Parisi SG, Andreoni M. HIV Replication at Low Copy Number and its Correlation with the HIV Reservoir: A Clinical Perspective. Curr HIV Res 2016; 13:250-7. [PMID: 25845389 PMCID: PMC4460281 DOI: 10.2174/1570162x13666150407142539] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/27/2015] [Accepted: 04/02/2015] [Indexed: 01/01/2023]
Abstract
The efficacy of combination therapy (antiretroviral therapy - ARV) is demonstrated by the high rates of viral suppression achieved in most treated HIV patients. Whereas contemporary
treatments may continuously suppress HIV replication, they do not eliminate the latent reservoir, which can reactivate HIV infection if ARV is discontinued. The persistence of HIV proviral DNA and
infectious viruses in CD4+ T cells and others cells has long been considered a major obstacle in eradicating the HIV virus in treated patients. Moreover, recent studies have demonstrated the
persistence of HIV replication at low copies in most patients on suppressive ARV. The source of this ‘residual viraemia’ and whether it declines over years of therapy remain unknown. Similarly, little is known regarding the biological
relationships between the HIV reservoir and viral replication at low copies. The question of whether this ‘residual viraemia’ represents active replication or the release of non-productive virus from the reservoir has not been adequately
resolved. From a clinical perspective, both the quantification of the HIV reservoir and the detection of low levels of replication in full-responder patients on prolonged ARV may provide important information regarding the effectiveness of treatment
and the eradication of HIV. To date, the monitoring of these two parameters has been conducted only for research purposes; the routine use of standardised tests procedure is lacking.
This review aims to assess the current data regarding the correlation between HIV replication at low copies and the HIV reservoir and to provide useful information for clinicians.
Collapse
Affiliation(s)
- Loredana Sarmati
- Clinical Infectious Diseases, Tor Vergata University, V. Montpellier 1, 00133, Roma, Italy.
| | | | | | | |
Collapse
|
36
|
Novel Assays for Measurement of Total Cell-Associated HIV-1 DNA and RNA. J Clin Microbiol 2016; 54:902-11. [PMID: 26763968 DOI: 10.1128/jcm.02904-15] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/05/2016] [Indexed: 01/26/2023] Open
Abstract
Although a number of PCR-based quantitative assays for measuring HIV-1 persistence during suppressive antiretroviral therapy (ART) have been reported, a simple, sensitive, reproducible method is needed for application to large clinical trials. We developed novel quantitative PCR assays for cell-associated (CA) HIV-1 DNA and RNA, targeting a highly conserved region in HIV-1pol, with sensitivities of 3 to 5 copies/1 million cells. We evaluated the performance characteristics of the assays using peripheral blood mononuclear cells (PBMCs) from 5 viremic patients and 20 patients receiving effective ART. Total and resting CD4(+)T cells were isolated from a subset of patients and tested for comparison with PBMCs. The estimated standard deviations including interassay variability and intra-assay variability of the assays were modest, i.e., 0.15 and 0.10 log10copies/10(6)PBMCs, respectively, for CA HIV-1 DNA and 0.40 and 0.19 log10copies/10(6)PBMCs for CA HIV-1 RNA. Testing of longitudinally obtained PBMC samples showed little variation for either viremic patients (median fold differences of 0.80 and 0.88 for CA HIV-1 DNA and RNA, respectively) or virologically suppressed patients (median fold differences of 1.14 and 0.97, respectively). CA HIV-1 DNA and RNA levels were strongly correlated (r= 0.77 to 1;P= 0.0001 to 0.037) for assays performed using PBMCs from different sources (phlebotomy versus leukapheresis) or using total or resting CD4(+)T cells purified by either bead selection or flow cytometric sorting. Their sensitivity, reproducibility, and broad applicability to small numbers of mononuclear cells make these assays useful for observational and interventional studies that examine longitudinal changes in the numbers of HIV-1-infected cells and their levels of transcription.
Collapse
|
37
|
Collier AC, Chun TW, Maenza J, Coombs RW, Tapia K, Chang M, Stevens CE, Justement JS, Murray D, Stekler JD, Mullins JI, Holte SE. A Pilot Study of Raltegravir Plus Combination Antiretroviral Therapy in Early Human Immunodeficiency Virus Infection: Challenges and Lessons Learned. Biores Open Access 2016; 5:15-21. [PMID: 26862469 PMCID: PMC4744890 DOI: 10.1089/biores.2015.0038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Availability of integrase strand transfer inhibitors created interest in determining whether their use would decrease persistently infected cell numbers. This study hypothesized that adding raltegravir (RAL) to standard antiretroviral therapy (ART) would decrease human immunodeficiency virus (HIV)-infected CD4+ T cells more than standard combination ART. This was a pilot, randomized study comparing open-label standard triple ART to standard triple ART plus RAL over 96 weeks in ART-naive adults with early HIV infection. The primary objective was to compare quantity and trajectory of HIV DNA. Eighty-two persons were referred. A diverse set of reasons precluded the enrollment of all but 10. Those who enrolled and completed the study had an estimated median duration of HIV infection of 74 days at ART start. The groups had similar baseline characteristics. The RAL group had more rapid first phase plasma HIV RNA decay (0.67 log10 copies/mL/day) than with combination ART (0.34 log10copies/mL/day), p = 0.037. Second phase HIV RNA decay, residual viremia, cell-associated RNA, HIV DNA, CD4+ T-cells with replication-competent virus, and 2LTR circle levels did not differ between groups. Among those with entry plasma HIV RNA levels above the median, 2LTR circles were significantly lower over time than in those with lower entry HIV RNA levels (p = 0.02). Our results suggest homogeneity of responses in cell-associated RNA, HIV DNA, CD4+ T-cells with replication-competent virus, and 2LTR circles with early HIV in both ART groups. The kinetics of 2LTR DNA did not reflect the kinetics of plasma HIV RNA decline following ART initiation.
Collapse
Affiliation(s)
- Ann C Collier
- Division of Infectious Diseases, Department of Medicine, University of Washington , Seattle, Washington
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland
| | - Janine Maenza
- Division of Infectious Diseases, Department of Medicine, University of Washington , Seattle, Washington
| | - Robert W Coombs
- Division of Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington.; Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Kenneth Tapia
- Department of Global Health, University of Washington , Seattle, Washington
| | - Ming Chang
- Department of Laboratory Medicine, University of Washington , Seattle, Washington
| | - Claire E Stevens
- Department of Neurology, University of Washington , Seattle, Washington
| | - J Shawn Justement
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland
| | - Danielle Murray
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland
| | - Joanne D Stekler
- Division of Infectious Diseases, Department of Medicine, University of Washington , Seattle, Washington
| | - James I Mullins
- Division of Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington.; Department of Laboratory Medicine, University of Washington, Seattle, Washington.; Department of Microbiology, University of Washington, Seattle, Washington
| | - Sarah E Holte
- Program in Biostatistics and Biomathematics, Division of Fred Hutch, Department of Biostatistics, University of Washington , Seattle, Washington
| |
Collapse
|
38
|
Vergnon-Miszczycha D, Lucht F, Roblin X, Pozzetto B, Paul S, Bourlet T. [Key role played by the gut associated lymphoid tissue during human immunodeficiency virus infection]. Med Sci (Paris) 2015; 31:1092-1101. [PMID: 26672662 DOI: 10.1051/medsci/20153112012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The gut associated lymphoid tissue (GALT) is the site of numerous immunological disturbances during HIV-1 infection. It constitutes the largest reservoir for HIV, not or very poorly susceptible to antiretroviral therapy (ART), making it a major obstacle to HIV cure. Moreover, the GALT is involved in systemic immune activation in HIV-infected individuals: intestinal damage due to viral replication and severe CD4(+) T cell depletion in the GALT leads to microbial translocation, a key driver of immune activation, and in turn, disease progression. In this review, we describe the role of the GALT in HIV infection and we discuss therapeutic options to decrease the intestinal viral reservoir and to preserve immune function in the gut of HIV-infected people. Achieving these goals is necessary for a long-term infection control after the interruption of ART.
Collapse
Affiliation(s)
- Delphine Vergnon-Miszczycha
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Service de maladies infectieuses et tropicales, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Frédéric Lucht
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Service de maladies infectieuses et tropicales, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Xavier Roblin
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Service de gastro-entérologie, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Bruno Pozzetto
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Laboratoire des agents infectieux et d'hygiène, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Stéphane Paul
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Laboratoire d'immunologie, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| | - Thomas Bourlet
- GIMAP/EA3064, Inserm CIC 1408 vaccinologie, université de Saint-Étienne, COMUE de Lyon, 42023 Saint-Étienne, France - Laboratoire des agents infectieux et d'hygiène, CHU de Saint-Étienne, 42055 Saint-Étienne Cedex 02, France
| |
Collapse
|
39
|
Rutstein SE, Sellers CJ, Ananworanich J, Cohen MS. The HIV treatment cascade in acutely infected people: informing global guidelines. Curr Opin HIV AIDS 2015; 10:395-402. [PMID: 26371460 PMCID: PMC4739850 DOI: 10.1097/coh.0000000000000193] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Acute and early HIV (AHI) is a pivotal time during HIV infection, yet there remain major shortfalls in diagnosis, linkage to care, and antiretroviral therapy (ART) initiation during AHI. We introduce an AHI-specific cascade, review recent evidence pertaining to the unique challenges of AHI, and discuss strategies for improving individual and public health outcomes. RECENT FINDINGS Presentation during AHI is common. Expanding use of fourth-generation testing and pooled nucleic acid amplification testing has led to improved AHI detection in resource-wealthy settings. Technologies capable of AHI diagnosis are rare in resource-limited settings; further development of point-of-care devices and utilization of targeted screening is needed. Rapid ART initiation during AHI limits reservoir seeding, preserves immunity, and prevents transmission. Reporting of AHI cascade outcomes is limited, but new evidence suggests that impressive rates of diagnosis, linkage to care, rapid ART initiation, and viral suppression can be achieved. SUMMARY With advancements in AHI diagnostics and strong evidence for the therapeutic and prevention benefits of ART initiated during AHI, improving AHI cascade outcomes is both crucial and feasible. HIV guidelines should recommend diagnostic algorithms capable of detecting AHI and prescribe rapid, universal ART initiation during AHI.
Collapse
Affiliation(s)
- Sarah E. Rutstein
- Department of Health Policy and Management, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Christopher J. Sellers
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jintanat Ananworanich
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Myron S. Cohen
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
40
|
Novel RNA Duplex Locks HIV-1 in a Latent State via Chromatin-mediated Transcriptional Silencing. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e261. [PMID: 26506039 PMCID: PMC4881759 DOI: 10.1038/mtna.2015.31] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/17/2015] [Indexed: 11/18/2022]
Abstract
Transcriptional gene silencing (TGS) of mammalian genes can be induced by short interfering RNA (siRNA) targeting promoter regions. We previously reported potent TGS of HIV-1 by siRNA (PromA), which targets tandem NF-κB motifs within the viral 5′LTR. In this study, we screened a siRNA panel with the aim of identifying novel 5′LTR targets, to provide multiplexing potential with enhanced viral silencing and application toward developing alternate therapeutic strategies. Systematic examination identified a novel siRNA target, si143, confirmed to induce TGS as the silencing mechanism. TGS was prolonged with virus suppression >12 days, despite a limited ability to induce post- TGS. Epigenetic changes associated with silencing were suggested by partial reversal by histone deacetylase inhibitors and confirmed by chromatin immunoprecipitation analyses, which showed induction of H3K27me3 and H3K9me3, reduction in H3K9Ac, and recruitment of argonaute-1, all characteristic marks of heterochromatin and TGS. Together, these epigenetic changes mimic those associated with HIV-1 latency. Further, robust resistance to reactivation was observed in the J-Lat 9.2 cell latency model, when transduced with shPromA and/or sh143. These data support si/shRNA-mediated TGS approaches to HIV-1 and provide alternate targets to pursue a functional cure, whereby the viral reservoir is locked in latency following antiretroviral therapy cessation.
Collapse
|
41
|
Colineau L, Rouers A, Yamamoto T, Xu Y, Urrutia A, Pham HP, Cardinaud S, Samri A, Dorgham K, Coulon PG, Cheynier R, Hosmalin A, Oksenhendler E, Six A, Kelleher AD, Zaunders J, Koup RA, Autran B, Moris A, Graff-Dubois S. HIV-Infected Spleens Present Altered Follicular Helper T Cell (Tfh) Subsets and Skewed B Cell Maturation. PLoS One 2015; 10:e0140978. [PMID: 26501424 PMCID: PMC4621058 DOI: 10.1371/journal.pone.0140978] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/16/2015] [Indexed: 12/27/2022] Open
Abstract
Follicular helper T (Tfh) cells within secondary lymphoid organs control multiple steps of B cell maturation and antibody (Ab) production. HIV-1 infection is associated with an altered B cell differentiation and Tfh isolated from lymph nodes of HIV-infected (HIV+) individuals provide inadequate B cell help in vitro. However, the mechanisms underlying this impairment of Tfh function are not fully defined. Using a unique collection of splenocytes, we compared the frequency, phenotype and transcriptome of Tfh subsets in spleens from HIV negative (HIV-) and HIV+ subjects. We observed an increase of CXCR5+PD-1highCD57-Tfh and germinal center (GC) CD57+ Tfh in HIV+ spleens. Both subsets showed a reduced mRNA expression of the transcription factor STAT-3, co-stimulatory, regulatory and signal transduction molecules as compared to HIV- spleens. Similarly, Foxp3 expressing follicular regulatory T (Tfr) cells were increased, suggesting sustained GC reactions in chronically HIV+ spleens. As a consequence, GC B cell populations were expanded, however, complete maturation into memory B cells was reduced in HIV+ spleens where we evidenced a compromised production of B cell-activating cytokines such as IL-4 and IL-10. Collectively our data indicate that, although Tfh proliferation and GC reactions seem to be ongoing in HIV-infected spleens, Tfh “differentiation” and expression of costimulatory molecules is skewed with a profound effect on B cell maturation.
Collapse
Affiliation(s)
- Lucie Colineau
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- CNRS, ERL 8255, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
| | - Angeline Rouers
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- CNRS, ERL 8255, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
| | - Takuya Yamamoto
- Immunology Laboratory, Vaccine research center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, United States of America
| | - Yin Xu
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
| | - Alejandra Urrutia
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- CNRS, ERL 8255, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
| | - Hang-Phuong Pham
- Sorbonne Universités UPMC Université Paris 06, UMRS 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, France
- INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, France
- CNRS, FRE3632, Immunology-Immunopathology-Immunotherapy (I3), Paris, France
| | - Sylvain Cardinaud
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- CNRS, ERL 8255, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
| | - Assia Samri
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- AP-HP, Hôpital Pitié-Salpêtière, Department of Immunology, Paris, France
| | - Karim Dorgham
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- AP-HP, Hôpital Pitié-Salpêtière, Department of Immunology, Paris, France
| | - Pierre-Grégoire Coulon
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- CNRS, ERL 8255, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
| | - Rémi Cheynier
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Hosmalin
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- AP-HP, Hôpital Cochin, Paris, France
| | - Eric Oksenhendler
- Université Paris Diderot, Assistance Publique-Hôpitaux de Paris, Département d’Immunologie Clinique, Hôpital Saint-Louis, Paris, France
| | - Adrien Six
- Sorbonne Universités UPMC Université Paris 06, UMRS 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, France
- INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, France
- CNRS, FRE3632, Immunology-Immunopathology-Immunotherapy (I3), Paris, France
| | - Anthony D. Kelleher
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
| | - John Zaunders
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney, Australia
| | - Richard A. Koup
- Immunology Laboratory, Vaccine research center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, United States of America
| | - Brigitte Autran
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- AP-HP, Hôpital Pitié-Salpêtière, Department of Immunology, Paris, France
| | - Arnaud Moris
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- CNRS, ERL 8255, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- AP-HP, Hôpital Pitié-Salpêtière, Department of Immunology, Paris, France
| | - Stéphanie Graff-Dubois
- Sorbonne Universités, UPMC Université Paris 06, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- INSERM, U1135, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- CNRS, ERL 8255, Center for Immunology and Microbial Infections—CIMI-Paris, Paris, France
- * E-mail:
| |
Collapse
|
42
|
Søgaard OS, Graversen ME, Leth S, Olesen R, Brinkmann CR, Nissen SK, Kjaer AS, Schleimann MH, Denton PW, Hey-Cunningham WJ, Koelsch KK, Pantaleo G, Krogsgaard K, Sommerfelt M, Fromentin R, Chomont N, Rasmussen TA, Østergaard L, Tolstrup M. The Depsipeptide Romidepsin Reverses HIV-1 Latency In Vivo. PLoS Pathog 2015; 11:e1005142. [PMID: 26379282 PMCID: PMC4575032 DOI: 10.1371/journal.ppat.1005142] [Citation(s) in RCA: 434] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/11/2015] [Indexed: 02/06/2023] Open
Abstract
Pharmacologically-induced activation of replication competent proviruses from latency in the presence of antiretroviral treatment (ART) has been proposed as a step towards curing HIV-1 infection. However, until now, approaches to reverse HIV-1 latency in humans have yielded mixed results. Here, we report a proof-of-concept phase Ib/IIa trial where 6 aviremic HIV-1 infected adults received intravenous 5 mg/m2 romidepsin (Celgene) once weekly for 3 weeks while maintaining ART. Lymphocyte histone H3 acetylation, a cellular measure of the pharmacodynamic response to romidepsin, increased rapidly (maximum fold range: 3.7–7.7 relative to baseline) within the first hours following each romidepsin administration. Concurrently, HIV-1 transcription quantified as copies of cell-associated un-spliced HIV-1 RNA increased significantly from baseline during treatment (range of fold-increase: 2.4–5.0; p = 0.03). Plasma HIV-1 RNA increased from <20 copies/mL at baseline to readily quantifiable levels at multiple post-infusion time-points in 5 of 6 patients (range 46–103 copies/mL following the second infusion, p = 0.04). Importantly, romidepsin did not decrease the number of HIV-specific T cells or inhibit T cell cytokine production. Adverse events (all grade 1–2) were consistent with the known side effects of romidepsin. In conclusion, romidepsin safely induced HIV-1 transcription resulting in plasma HIV-1 RNA that was readily detected with standard commercial assays demonstrating that significant reversal of HIV-1 latency in vivo is possible without blunting T cell-mediated immune responses. These finding have major implications for future trials aiming to eradicate the HIV-1 reservoir. One proposed way of curing HIV is to activate virus transcription and kill latently infected cells while the presence of antiretroviral therapy prevents spreading the infection. Induction of global T cell activation by mitogenic or other potent activators effectively reverses HIV-1 from latency ex vivo, but such compounds are generally too toxic for clinical use. Therefore, investigating the capacity of small molecule latency reversing agents to induce production of virus without causing global T cell activation has been a top research priority for scientists in recent years. In the present clinical trial, we demonstrate that significant viral reactivation can be safely induced using the depsipeptide romidepsin (HDAC inhibitor) in long-term suppressed HIV-1 individuals on antiretroviral therapy. Following each romidepsin infusion, we observed clear increases in lymphocyte H3 acetylation, HIV-1 transcription, and plasma HIV-1 RNA. Importantly, this reversal of HIV-1 latency could be measured using standard clinical assays for detection of plasma HIV-1 RNA. Furthermore, romidepsin did not alter the proportion of HIV-specific T cells or inhibit T cell cytokine production which is critically important for future trials combining HDAC inhibitors with interventions (e.g. therapeutic HIV-1 vaccination) designed to enhance killing of latently infected cells.
Collapse
Affiliation(s)
- Ole S. Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- * E-mail:
| | - Mette E. Graversen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Steffen Leth
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rikke Olesen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | | - Sara K. Nissen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Sofie Kjaer
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mariane H. Schleimann
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Paul W. Denton
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Aarhus Institute for Advanced Studies, Aarhus University, Denmark
| | - William J. Hey-Cunningham
- Kirby Institute, University of New South Wales Medicine, University of New South Wales Australia, Sydney, Australia
| | - Kersten K. Koelsch
- Kirby Institute, University of New South Wales Medicine, University of New South Wales Australia, Sydney, Australia
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | | | - Nicolas Chomont
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, Montreal, Quebec, Canada
| | - Thomas A. Rasmussen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
43
|
Ahlenstiel CL, Suzuki K, Marks K, Symonds GP, Kelleher AD. Controlling HIV-1: Non-Coding RNA Gene Therapy Approaches to a Functional Cure. Front Immunol 2015; 6:474. [PMID: 26441979 PMCID: PMC4584958 DOI: 10.3389/fimmu.2015.00474] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 08/31/2015] [Indexed: 12/27/2022] Open
Abstract
The current treatment strategy for HIV-1 involves prolonged and intensive combined antiretroviral therapy (cART), which successfully suppresses plasma viremia. It has transformed HIV-1 infection into a chronic disease. However, despite the success of cART, a latent form of HIV-1 infection persists as integrated provirus in resting memory CD4(+) T cells. Virus can reactivate from this reservoir upon cessation of treatment, and hence HIV requires lifelong therapy. The reservoir represents a major barrier to eradication. Understanding molecular mechanisms regulating HIV-1 transcription and latency are crucial to develop alternate treatment strategies, which impact upon the reservoir and provide a path toward a "functional cure" in which there is no detectable viremia in the absence of cART. Numerous reports have suggested ncRNAs are involved in regulating viral transcription and latency. This review will discuss the latest developments in ncRNAs, specifically short interfering (si)RNA and short hairpin (sh)RNA, targeting molecular mechanisms of HIV-1 transcription, which may represent potential future therapeutics. It will also briefly address animal models for testing potential therapeutics and current gene therapy clinical trials.
Collapse
Affiliation(s)
| | - Kazuo Suzuki
- The Kirby Institute, UNSW Australia, Sydney, NSW, Australia
- Immunovirology Laboratory, St. Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | - Katherine Marks
- Immunovirology Laboratory, St. Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | | | - Anthony D. Kelleher
- The Kirby Institute, UNSW Australia, Sydney, NSW, Australia
- Immunovirology Laboratory, St. Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
44
|
How does the timing of antiretroviral therapy initiation in acute infection affect HIV reservoirs? Curr Opin HIV AIDS 2015; 10:18-28. [PMID: 25415421 DOI: 10.1097/coh.0000000000000122] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW The long-lived viral reservoir is a major obstacle to achieving a cure for HIV. Therapeutic strategies, such as early antiretroviral therapy (ART), may be a prerequisite to achieving long-term control of viral replication upon ART withdrawal. RECENT FINDINGS HIV persistence is established early in acute HIV infection (AHI) with infection in long-lived memory CD4⁺ T cells. Studies conducted in nonhuman primates have suggested that this could occur as early as 3 days postinfection; however, the timing in humans is uncertain. ART during AHI significantly restricts the HIV reservoirs as compared with later treatment. Early ART, particularly prior to the detection of HIV immunoglobulin M, may also reduce the contribution of the long-lived central memory CD4⁺ T cells to the total HIV reservoir, a profile observed in individuals who naturally control HIV without ART. SUMMARY It is clear that early ART has a greater impact in limiting the HIV reservoirs than later treatment. However, latently infected long-lived memory CD4⁺ T cells persist in most early treated individuals. Therefore, additional interventions will likely be required to eliminate all cells capable of producing replication-competent virus but treatment in AHI may be the critical first step in containing the HIV reservoirs.
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW To review current knowledge about the impact of long-term combination antiretroviral therapy (cART) on HIV reservoirs. RECENT FINDINGS The number of HIV-infected cells that persist during long-term antiretroviral therapy is associated with the stage of HIV infection at the time of treatment initiation. Initiation of cART reduces the number of infected cells over the first 4 years of therapy, but thereafter there is no further decline despite long-term effective cART. The remarkable stability of infected cell numbers is likely due to a balance among homeostatic or antigen-driven proliferation of infected memory T-cells subsets, clonal expansion of a subset of infected cells as a consequence of specific retroviral integration sites, and death of other infected cells. At present, there is no effective means of accelerating the decay of infected cells in individuals initiated on cART during chronic HIV infection. SUMMARY Given the stability and difficulty in eliminating HIV-infected cells, early initiation of cART in treatment-naïve HIV-infected patients is currently the most effective way to limit the size and diversity of HIV reservoirs.
Collapse
|
46
|
Immunological and pharmacological strategies to reactivate HIV-1 from latently infected cells: a possibility for HIV-1 paediatric patients? J Virus Erad 2015. [DOI: 10.1016/s2055-6640(20)30508-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
47
|
Pinkevych M, Cromer D, Tolstrup M, Grimm AJ, Cooper DA, Lewin SR, Søgaard OS, Rasmussen TA, Kent SJ, Kelleher AD, Davenport MP. HIV Reactivation from Latency after Treatment Interruption Occurs on Average Every 5-8 Days--Implications for HIV Remission. PLoS Pathog 2015; 11:e1005000. [PMID: 26133551 PMCID: PMC4489624 DOI: 10.1371/journal.ppat.1005000] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/03/2015] [Indexed: 12/11/2022] Open
Abstract
HIV infection can be effectively controlled by anti-retroviral therapy (ART) in most patients. However therapy must be continued for life, because interruption of ART leads to rapid recrudescence of infection from long-lived latently infected cells. A number of approaches are currently being developed to 'purge' the reservoir of latently infected cells in order to either eliminate infection completely, or significantly delay the time to viral recrudescence after therapy interruption. A fundamental question in HIV research is how frequently the virus reactivates from latency, and thus how much the reservoir might need to be reduced to produce a prolonged antiretroviral-free HIV remission. Here we provide the first direct estimates of the frequency of viral recrudescence after ART interruption, combining data from four independent cohorts of patients undergoing treatment interruption, comprising 100 patients in total. We estimate that viral replication is initiated on average once every ≈6 days (range 5.1- 7.6 days). This rate is around 24 times lower than previous thought, and is very similar across the cohorts. In addition, we analyse data on the ratios of different 'reactivation founder' viruses in a separate cohort of patients undergoing ART-interruption, and estimate the frequency of successful reactivation to be once every 3.6 days. This suggests that a reduction in the reservoir size of around 50-70-fold would be required to increase the average time-to-recrudescence to about one year, and thus achieve at least a short period of anti-retroviral free HIV remission. Our analyses suggests that time-to-recrudescence studies will need to be large in order to detect modest changes in the reservoir, and that macaque models of SIV latency may have much higher frequencies of viral recrudescence after ART interruption than seen in human HIV infection. Understanding the mean frequency of recrudescence from latency is an important first step in approaches to prolong antiretroviral-free viral remission in HIV.
Collapse
Affiliation(s)
- Mykola Pinkevych
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Deborah Cromer
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Andrew J. Grimm
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - David A. Cooper
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Sharon R. Lewin
- Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Ole S. Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas A. Rasmussen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | - Miles P. Davenport
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| |
Collapse
|
48
|
Intensive five-drug antiretroviral therapy regimen versus standard triple-drug therapy during primary HIV-1 infection (OPTIPRIM-ANRS 147): a randomised, open-label, phase 3 trial. THE LANCET. INFECTIOUS DISEASES 2015; 15:387-96. [PMID: 25701561 DOI: 10.1016/s1473-3099(15)70021-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Early combination antiretroviral therapy (cART) initiation at the time of primary HIV-1 infection could restrict the establishment of HIV reservoirs. We aimed to assess the effect of a cART regimen intensified with raltegravir and maraviroc, compared with standard triple-drug cART, on HIV-DNA load. METHODS In this randomised, open-label, phase 3 trial, we recruited patients from hospitals across France. Inclusion criteria were primary HIV-1 infection (an incomplete HIV-1 western blot and detectable plasma HIV-RNA), with either symptoms or a CD4+ cell count below 500 cells per μL. Patients were randomly assigned (1:1) to an intensive, five-drug cART regimen (raltegravir 400 mg and maraviroc 150 mg twice daily, and a fixed-dose combination of tenofovir disoproxil fumarate 300 g plus emtricitabine 200 g, darunavir 800 g, and ritonavir 100 g once daily) or a standard triple-drug cART regimen (tenofovir disoproxil fumarate 300 g plus emtricitabine 200 g, darunavir 800 g, and ritonavir 100 g once daily) using a predefined randomised list generated by randomly selected variable block sizes. The primary endpoint was the median number of HIV-DNA copies per 10(6) peripheral blood mononuclear cells (PBMC) at month 24, analysed in the modified intention-to-treat population, defined as all patients who started their assigned treatment. This study is registered with ClinicalTrials.gov, number NCT01033760. FINDINGS Between April 26, 2010, and July 13, 2011, 110 patients were enrolled, of whom 92 were randomly assigned and 90 started treatment (45 in each treatment group). Six (13%) patients in the intensive cART group and two (4%) in the standard cART group discontinued before month 24. At month 24, HIV-DNA loads were similar between groups (2·35 [IQR 2·05-2·50] log₁₀ per 10(6) PBMC in the intensive cART group vs 2·25 [1·71-2·55] in the standard cART group; p=0·21). Eight grade 3-4 clinical adverse events were reported in seven patients in the intensive cART group and seven grade 3-4 clinical adverse events were reported in seven patients in the standard cART group. Three serious clinical adverse events occurred: two (pancreatitis and lipodystrophy) in the standard cART group, which were regarded as treatment related, and one event (suicide attempt) in the intensive cART group that was unrelated to treatment. INTERPRETATION After 24 months, cART intensified with raltegravir and maraviroc did not have a greater effect on HIV blood reservoirs than did standard cART. These results should help to design future trials of treatments aiming to decrease the HIV reservoir in patients with primary HIV-1 infection. FUNDING Inserm-ANRS, Gilead Sciences, Janssen Pharmaceuticals, Merck, and ViiV Laboratories.
Collapse
|
49
|
Dynamics of HIV DNA and Residual Viremia in Patients Treated With a Raltegravir-Containing Regimen. J Acquir Immune Defic Syndr 2015; 68:e18-20. [DOI: 10.1097/qai.0000000000000414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
50
|
Evaluation of the effect of short-term treatment with the integrase inhibitor raltegravir (Isentress) on the course of progressive feline leukemia virus infection. Vet Microbiol 2014; 175:167-78. [PMID: 25500005 DOI: 10.1016/j.vetmic.2014.10.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 11/22/2022]
Abstract
Cats persistently infected with the gammaretrovirus feline leukemia virus (FeLV) are at risk to die within months to years from FeLV-associated disease, such as immunosuppression, anemia or lymphoma/leukemia. The integrase inhibitor raltegravir has been demonstrated to reduce FeLV replication in vitro. The aim of the present study was to investigate raltegravir in vivo for its safety and efficacy to suppress FeLV replication. The safety was tested in three naïve specified pathogen-free (SPF) cats during a 15 weeks treatment period (initially 20mg then 40mg orally b.i.d.). No adverse effects were noted. The efficacy was tested in seven persistently FeLV-infected SPF cats attained from 18 cats experimentally exposed to FeLV-A/Glasgow-1. The seven cats were treated during nine weeks (40mg then 80mg b.i.d.). Raltegravir was well tolerated even at the higher dose. A significant decrease in plasma viral RNA loads (∼5×) was found; however, after treatment termination a rebound effect was observed. Only one cat developed anti-FeLV antibodies and viral RNA loads remained decreased after treatment termination. Of note, one of the untreated FeLV-A infected cats developed fatal FeLV-C associated anemia within 5 weeks of FeLV-A infection. Moreover, progressive FeLV infection was associated with significantly lower enFeLV loads prior to infection supporting that FeLV susceptibility may be related to the genetic background of the cat. Overall, our data demonstrate the ability of raltegravir to reduce viral replication also in vivo. However, no complete control of viremia was achieved. Further investigations are needed to find an optimized treatment against FeLV. (250 words).
Collapse
|