1
|
Gálvez C, Urrea V, Garcia-Guerrero MDC, Bernal S, Benet S, Mothe B, Bailón L, Dalmau J, Martinez A, Nieto A, Leal L, García F, Clotet B, Martinez-Picado J, Salgado M. Altered T-cell subset distribution in the viral reservoir in HIV-1-infected individuals with extremely low proviral DNA (LoViReTs). J Intern Med 2022; 292:308-320. [PMID: 35342993 PMCID: PMC9308636 DOI: 10.1111/joim.13484] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND HIV cure strategies aim to eliminate viral reservoirs that persist despite successful antiretroviral therapy (ART). We have previously described that 9% of HIV-infected individuals who receive ART harbor low levels of provirus (LoViReTs). METHODS We selected 22 LoViReTs matched with 22 controls ART suppressed for more than 3 years with fewer than 100 and more than 100 HIV-DNA copies/106 CD4+ T cells, respectively. We measured HIV reservoirs in blood and host genetic factors. Fourteen LoViReTs underwent leukapheresis to analyze replication-competent virus, and HIV-DNA in CD4+ T-cell subpopulations. Additionally, we measured HIV-DNA in rectum and/or lymph node biopsies from nine of them. RESULTS We found that LoViReTs harbored not only lower levels of total HIV-DNA, but also significantly lower intact HIV-DNA, cell-associated HIV-RNA, and ultrasensitive viral load than controls. The proportion of intact versus total proviruses was similar in both groups. We found no differences in the percentage of host factors. In peripheral blood, 71% of LoViReTs had undetectable replication-competent virus. Minimum levels of total HIV-DNA were found in rectal and lymph node biopsies compared with HIV-infected individuals receiving ART. The main contributors to the reservoir were short-lived transitional memory and effector memory T cells (47% and 29%, respectively), indicating an altered distribution of the HIV reservoir in the peripheral T-cell subpopulations of LoViReTs. CONCLUSION In conclusion, LoViReTs are characterized by low levels of viral reservoir in peripheral blood and secondary lymphoid tissues, which might be explained by an altered distribution of the proviral HIV-DNA towards more short-lived memory T cells. LoViReTs can be considered exceptional candidates for future interventions aimed at curing HIV.
Collapse
Affiliation(s)
- Cristina Gálvez
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain
| | - Víctor Urrea
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain
| | - Maria Del Carmen Garcia-Guerrero
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain
| | - Sílvia Bernal
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain.,Chair in Infectious Diseases and Immunity, University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Susana Benet
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain.,Lluita contra la SIDA Foundation, Infectious Diseases Department, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain.,Chair in Infectious Diseases and Immunity, University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain.,Lluita contra la SIDA Foundation, Infectious Diseases Department, Hospital Germans Trias i Pujol, Badalona, Spain.,CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Lucía Bailón
- Lluita contra la SIDA Foundation, Infectious Diseases Department, Hospital Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, Autonomous University of Barcelona, Catalonia, Spain
| | - Judith Dalmau
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain
| | - Andrea Martinez
- Lluita contra la SIDA Foundation, Infectious Diseases Department, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Aroa Nieto
- Lluita contra la SIDA Foundation, Infectious Diseases Department, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Lorna Leal
- Infectious Diseases Department Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Felipe García
- Infectious Diseases Department Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain.,Chair in Infectious Diseases and Immunity, University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain.,Lluita contra la SIDA Foundation, Infectious Diseases Department, Hospital Germans Trias i Pujol, Badalona, Spain.,CIBER de Enfermedades Infecciosas, Madrid, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain.,Chair in Infectious Diseases and Immunity, University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain.,CIBER de Enfermedades Infecciosas, Madrid, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Maria Salgado
- IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Badalona, Spain.,CIBER de Enfermedades Infecciosas, Madrid, Spain
| |
Collapse
|
2
|
Atlas of the HIV-1 Reservoir in Peripheral CD4 T Cells of Individuals on Successful Antiretroviral Therapy. mBio 2021; 12:e0307821. [PMID: 34844430 PMCID: PMC8630536 DOI: 10.1128/mbio.03078-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Knowing the mechanisms that govern the persistence of infected CD4+ subpopulations could help us to design new therapies to cure HIV-1 infection. We evaluated the simultaneous distribution of the HIV-1 reservoir in 13 CD4+ subpopulations from 14 HIV-1-infected individuals on antiretroviral therapy to analyze its relationship with HIV-1 transcription, immune activation, and cell proliferation. A unique large blood donation was used to isolate CD4, CD4 resting (CD4r), CD4 activated (CD4a), T naive (TN), T stem cell memory (TSCM), T central memory (TCM), T transitional memory (TTM), T effector memory (TEM), circulating T follicular helper (cTFH), TCD20, TCD32, and resting memory TCD2high (rmTCD2high) cells. HIV-1 DNA measured by droplet digital PCR ranged from 3,636 copies/106 in TTM to 244 in peripheral blood mononuclear cells (PBMCs), with no subpopulation standing out for provirus enrichment. Importantly, all the subpopulations harbored intact provirus by intact provirus DNA assay (IPDA). TCD32, cTFH, and TTM had the highest levels of HIV-1 transcription measured by fluorescent in situ hybridization with flow cytometry (FISH/flow), but without reaching statistical differences. The subpopulations more enriched in provirus had a memory phenotype, were less activated (measured by CD38+/HLA-DR+), and expressed more programmed cell death 1 (PD-1). Conversely, subpopulations transcribing more HIV-1 RNA were not necessarily enriched in provirus and were more activated (measured by CD38+/HLA-DR+) and more proliferative (measured by Ki-67). In conclusion, the HIV reservoir is composed of a mosaic of subpopulations contributing to the HIV-1 persistence through different mechanisms such as susceptibility to infection, provirus intactness, or transcriptional status. The narrow range of reservoir differences between the different blood cell subsets tested suggests limited efficacy in targeting only specific cell subpopulations during HIV-1 cure strategies. IMPORTANCE The main barrier for HIV-1 cure is the presence of latently infected CD4+ T cells. Although various cell subpopulations have been identified as major HIV-1 reservoir cells, the relative contribution of infected CD4 subpopulations in the HIV-1 reservoir remains largely unknown. Here, we evaluated the simultaneous distribution of the HIV-1 reservoir in 13 CD4+ T-cell subpopulations in peripheral blood from HIV-1-infected individuals under suppressive antiretroviral therapy. We found that the HIV-1 reservoir is composed of a mosaic of cell subpopulations, with heterogeneous proviral DNA, HIV-1 transcription, and activation status. Hence, each cell subpopulation contributes to the HIV-1 persistence through different mechanisms such as susceptibility to infection, rates of intact provirus, transcriptional status or half-life. This research provides new insights into the composition of the HIV-1 reservoir, suggesting that, to be effective, eradication strategies must simultaneously target multiple cell subpopulations.
Collapse
|
3
|
Eberhard JM, Angin M, Passaes C, Salgado M, Monceaux V, Knops E, Kobbe G, Jensen B, Christopeit M, Kröger N, Vandekerckhove L, Badiola J, Bandera A, Raj K, van Lunzen J, Hütter G, Kuball JHE, Martinez-Laperche C, Balsalobre P, Kwon M, Díez-Martín JL, Nijhuis M, Wensing A, Martinez-Picado J, Schulze Zur Wiesch J, Sáez-Cirión A. Vulnerability to reservoir reseeding due to high immune activation after allogeneic hematopoietic stem cell transplantation in individuals with HIV-1. Sci Transl Med 2021; 12:12/542/eaay9355. [PMID: 32376772 DOI: 10.1126/scitranslmed.aay9355] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only medical intervention that has led to an HIV cure. Whereas the HIV reservoir sharply decreases after allo-HSCT, the dynamics of the T cell reconstitution has not been comprehensively described. We analyzed the activation and differentiation of CD4+ and CD8+ T cells, and the breadth and quality of HIV- and CMV-specific CD8+ T cell responses in 16 patients with HIV who underwent allo-HSCT (including five individuals who received cells from CCR5Δ32/Δ32 donors) to treat their underlying hematological malignancy and who remained on antiretroviral therapy (ART). We found that reconstitution of the T cell compartment after allo-HSCT was slow and heterogeneous with an initial expansion of activated CD4+ T cells that preceded the expansion of CD8+ T cells. Although HIV-specific CD8+ T cells disappeared immediately after allo-HSCT, weak HIV-specific CD8+ T cell responses were detectable several weeks after transplant and could still be detected at the time of full T cell chimerism, indicating that de novo priming, and hence antigen exposure, occurred during the time of T cell expansion. These HIV-specific T cells had limited functionality compared with CMV-specific CD8+ T cells and persisted years after allo-HSCT. In conclusion, immune reconstitution was slow, heterogeneous, and incomplete and coincided with de novo detection of weak HIV-specific T cell responses. The initial short phase of high T cell activation, in which HIV antigens were present, may constitute a window of vulnerability for the reseeding of viral reservoirs, emphasizing the importance of maintaining ART directly after allo-HSCT.
Collapse
Affiliation(s)
- Johanna M Eberhard
- 1. Department of Medicine, Infectious Diseases Unit, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany.,DZIF Partner Site (German Center for Infection Research), Hamburg-Lübeck-Borstel-Riems Site, Hamburg, Germany
| | - Mathieu Angin
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France
| | - Caroline Passaes
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France
| | - Maria Salgado
- AIDS Research Institute IrsiCaixa, 08916 Badalona, Spain
| | - Valerie Monceaux
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France
| | - Elena Knops
- Institute of Virology, University of Cologne, 50935 Cologne, Germany
| | - Guido Kobbe
- Department of Haematology, Oncology, and Clinical Immunology, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Björn Jensen
- Department of Gastroenterology, Hepatology, and Infectious Diseases, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center HamburgEppendorf, 20246 Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center HamburgEppendorf, 20246 Hamburg, Germany
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, B-9000 Ghent, Belgium
| | - Jon Badiola
- Hematology Department, Virgen de las Nieves University Hospital, 18014 Granada, Spain
| | | | - Kavita Raj
- Department of Haematology, King's College Hospital, London SE5 9RS, UK
| | - Jan van Lunzen
- 1. Department of Medicine, Infectious Diseases Unit, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany.,ViiV Healthcare, Brentford, Middlesex TW8 9GS, UK
| | | | | | - Carolina Martinez-Laperche
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - Pascual Balsalobre
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - Mi Kwon
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - José L Díez-Martín
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - Monique Nijhuis
- University Medical Center Utrecht, 3584 CX, Utrecht, Netherlands
| | | | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, 08916 Badalona, Spain.,UVic-UCC, 08500 Vic, Spain.,ICREA, 08010 Barcelona, Spain
| | - Julian Schulze Zur Wiesch
- 1. Department of Medicine, Infectious Diseases Unit, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany. .,DZIF Partner Site (German Center for Infection Research), Hamburg-Lübeck-Borstel-Riems Site, Hamburg, Germany
| | - Asier Sáez-Cirión
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France.
| |
Collapse
|
4
|
Casado C, Galvez C, Pernas M, Tarancon-Diez L, Rodriguez C, Sanchez-Merino V, Vera M, Olivares I, De Pablo-Bernal R, Merino-Mansilla A, Del Romero J, Lorenzo-Redondo R, Ruiz-Mateos E, Salgado M, Martinez-Picado J, Lopez-Galindez C. Permanent control of HIV-1 pathogenesis in exceptional elite controllers: a model of spontaneous cure. Sci Rep 2020; 10:1902. [PMID: 32024974 PMCID: PMC7002478 DOI: 10.1038/s41598-020-58696-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/27/2019] [Indexed: 11/09/2022] Open
Abstract
Elite controllers (EC) represent a small subset of HIV-1-infected people that spontaneously control viral replication. However, natural virological suppression and absence of immune dysfunction are not always long-term sustained. We define exceptional EC (EEC) as HIV-1 subjects who maintain the EC characteristics without disease progression for more than 25 years. We analyzed three EEC, diagnosed between 1988 and 1992, who never showed signs of clinical disease progression in absence of any antiretroviral treatment. A comprehensive clinical, virological, and immunological study was performed. The individuals simultaneously exhibited ≥3 described host protective alleles, low levels of total HIV-1 DNA (<20 copies/106 CD4+ T-cells) without evidence of replication-competent viruses (<0.025 IUPM), consistent with high levels of defective genomes, strong cellular HIV-1-specific immune response, and a high poly-functionality index (>0.50). Inflammation levels of EEC were similar to HIV-1 negative donors. Remarkably, they showed an exceptional lack of viral evolution and 8-fold lower genetic diversity (<0.01 s/n) in env gene than other EC. We postulate that these EEC represent cases of spontaneous functional HIV-1 cure. A non-functional and non-genetically evolving viral reservoir along with an HIV-1-specific immune response seems to be key for the spontaneous functional cure.
Collapse
Affiliation(s)
- Concepcion Casado
- Virología Molecular, Laboratorio de Referencia e Investigación en Retrovirus, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Cristina Galvez
- AIDS Research Institute IrsiCaixa, Badalona, Spain
- Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Maria Pernas
- Virología Molecular, Laboratorio de Referencia e Investigación en Retrovirus, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Laura Tarancon-Diez
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Carmen Rodriguez
- Centro Sanitario Sandoval, Hospital Clínico San Carlos. IdISSC, Madrid, Spain
| | - Víctor Sanchez-Merino
- AIDS Immunopathology Unit. Laboratorio de Referencia e Investigación en Retrovirus. Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Mar Vera
- Centro Sanitario Sandoval, Hospital Clínico San Carlos. IdISSC, Madrid, Spain
| | - Isabel Olivares
- Virología Molecular, Laboratorio de Referencia e Investigación en Retrovirus, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Rebeca De Pablo-Bernal
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Alberto Merino-Mansilla
- AIDS Immunopathology Unit. Laboratorio de Referencia e Investigación en Retrovirus. Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Jorge Del Romero
- Centro Sanitario Sandoval, Hospital Clínico San Carlos. IdISSC, Madrid, Spain
| | - Ramon Lorenzo-Redondo
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60011, USA
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | | | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, Badalona, Spain.
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| | - Cecilio Lopez-Galindez
- Virología Molecular, Laboratorio de Referencia e Investigación en Retrovirus, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
| |
Collapse
|
5
|
Blanch-Lombarte O, Gálvez C, Revollo B, Jiménez-Moyano E, Llibre JM, Manzano JL, Boada A, Dalmau J, E. Speiser D, Clotet B, G. Prado J, Martinez-Picado J. Enhancement of Antiviral CD8 + T-Cell Responses and Complete Remission of Metastatic Melanoma in an HIV-1-Infected Subject Treated with Pembrolizumab. J Clin Med 2019; 8:jcm8122089. [PMID: 31805700 PMCID: PMC6947580 DOI: 10.3390/jcm8122089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/14/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pembrolizumab is an immune checkpoint inhibitor against programmed cell death protein-1 (PD-1) approved for therapy in metastatic melanoma. PD-1 expression is associated with a diminished functionality in HIV-1 specific-CD8+ T cells. It is thought that PD-1 blockade could contribute to reinvigorate antiviral immunity and reduce the HIV-1 reservoir. METHODS Upon metastatic melanoma diagnosis, an HIV-1-infected individual on stable suppressive antiretroviral regimen was treated with pembrolizumab. A PET-CT was performed before and one year after pembrolizumab initiation. We monitored changes in the immunophenotype and HIV-1 specific-CD8+ T-cell responses during 36 weeks of treatment. Furthermore, we assessed changes in the viral reservoir by total HIV-1 DNA, cell-associated HIV-1 RNA, and ultrasensitive plasma viral load. RESULTS Complete metabolic response was achieved after pembrolizumab treatment of metastatic melanoma. Activated CD8+ T-cells expressing HLA-DR+/CD38+ transiently increased over the first nine weeks of treatment. Concomitantly, there was an augmented response of HIV-1 specific-CD8+ T cells with TNF production and poly-functionality, transitioning from TNF to an IL-2 profile. Furthermore, a transient reduction of 24% and 32% in total HIV-1 DNA was observed at weeks 3 and 27, respectively, without changes in other markers of viral persistence. CONCLUSIONS These data demonstrate that pembrolizumab may enhance the HIV-1 specific-CD8+ T-cell response, marginally affecting the HIV-1 reservoir. A transient increase of CD8+ T-cell activation, TNF production, and poly-functionality resulted from PD-1 blockade. However, the lack of sustained changes in the viral reservoir suggests that viral reactivation is needed concomitantly with HIV-1-specific immune enhancement.
Collapse
Affiliation(s)
- Oscar Blanch-Lombarte
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain; (O.B.-L.); (C.G.); (E.J.-M.); (J.D.); (B.C.)
- Autonomous University of Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain;
| | - Cristina Gálvez
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain; (O.B.-L.); (C.G.); (E.J.-M.); (J.D.); (B.C.)
- Autonomous University of Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain;
| | - Boris Revollo
- Infectious Diseases Department, University Hospital “Germans Trias i Pujol”, 08916 Badalona, Spain; (B.R.); (J.M.L.)
| | - Esther Jiménez-Moyano
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain; (O.B.-L.); (C.G.); (E.J.-M.); (J.D.); (B.C.)
| | - Josep M. Llibre
- Infectious Diseases Department, University Hospital “Germans Trias i Pujol”, 08916 Badalona, Spain; (B.R.); (J.M.L.)
| | - José Luís Manzano
- Medical Oncology Service—Badalona Applied Research Group in Oncology (B-ARGO Group), University Hospital “Germans Trias i Pujol”—Catalan Insitute of Oncology (ICO), 08916 Badalona, Spain;
| | - Aram Boada
- Autonomous University of Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain;
- Department of Dermatology, University Hospital “Germans Trias i Pujol”, 08916 Badalona, Spain
| | - Judith Dalmau
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain; (O.B.-L.); (C.G.); (E.J.-M.); (J.D.); (B.C.)
| | - Daniel E. Speiser
- Department of Oncology, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain; (O.B.-L.); (C.G.); (E.J.-M.); (J.D.); (B.C.)
- Autonomous University of Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain;
- Infectious Diseases Department, University Hospital “Germans Trias i Pujol”, 08916 Badalona, Spain; (B.R.); (J.M.L.)
- Chair in Infectious Diseases and Immunity, Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - Julia G. Prado
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain; (O.B.-L.); (C.G.); (E.J.-M.); (J.D.); (B.C.)
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Correspondence: (J.G.P.); (J.M.-P.)
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain; (O.B.-L.); (C.G.); (E.J.-M.); (J.D.); (B.C.)
- Chair in Infectious Diseases and Immunity, Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (J.G.P.); (J.M.-P.)
| |
Collapse
|
6
|
Agrati C, Tumino N, Bordoni V, Pinnetti C, Sabatini A, Amendola A, Abbate I, Lorenzini P, Mondi A, Casetti R, Cimini E, Grassi G, Antinori A, Sacchi A. Myeloid Derived Suppressor Cells Expansion Persists After Early ART and May Affect CD4 T Cell Recovery. Front Immunol 2019; 10:1886. [PMID: 31440256 PMCID: PMC6694843 DOI: 10.3389/fimmu.2019.01886] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/25/2019] [Indexed: 12/26/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are expanded during HIV-1 infection and correlated with disease progression. MDSC expand in the early phase of primary infection depending on TRAIL level. In this study we evaluated the effect of ART on the frequency of MDSC in patients with primary HIV infection (PHI), and their impact on CD4 T cell reconstitution. MDSC frequency was evaluated by flow-cytometry in 60 PHI patients at 12, 24 and 48 weeks after ART initiation. Cytokine plasma levels were evaluated by Luminex technology at the same time points. The capacity of MDSC to modulate hematopoietic early progenitor cells' expansion was evaluated using the OP9/Dl1 in vitro system. As previously described, polymorphonuclear-MDSC (PMN-MDSC) frequency was higher in PHI compared to healthy donors. Interestingly, 48 weeks of successful ART failed to normalize the PMN-MDSC frequency. Moreover, PMN-MDSC frequency was not correlated with residual viral load, suggesting that the persistence of PMN-MDSC was not due to residual viral replication. Interestingly, patients with low PMN-MDSC frequency (<6%) at T0 had a higher HIV DNA at the same time point than individuals with high PMN-MDSC frequency (>6%). We also found an inverse correlation between PMN-MDSC frequency and CD4-T cell count at 48 weeks post-ART, which was confirmed by multivariate analysis adjusting for age and CD4 T cell number at baseline. These data suggest that the persistence of PMN-MDSC may impact CD4 T cell recovery. Indeed, in vitro PMN-MDSC impaired the expansion of CD34+CD38- hematopoietic early progenitors. Further, a balance between TRAIL and GM-CSF may be necessary to maintain a low MDSC level. In conclusion, early ART initiation was not able to normalize PMN-MDSC frequency that might impact the CD4 T cell recovery. These data open new questions regarding the clinical impact of MDSC persistence in HIV+ patients, in particular on non-AIDS related diseases.
Collapse
Affiliation(s)
- Chiara Agrati
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Nicola Tumino
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Veronica Bordoni
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Carmela Pinnetti
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Sabatini
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Amendola
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Isabella Abbate
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Patrizia Lorenzini
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Annalisa Mondi
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Rita Casetti
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Germana Grassi
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Antinori
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Sacchi
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| |
Collapse
|
7
|
Gupta RK, Abdul-Jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, Martinez-Picado J, Nijhuis M, Wensing AMJ, Lee H, Grant P, Nastouli E, Lambert J, Pace M, Salasc F, Monit C, Innes AJ, Muir L, Waters L, Frater J, Lever AML, Edwards SG, Gabriel IH, Olavarria E. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature 2019; 568:244-248. [PMID: 30836379 PMCID: PMC7275870 DOI: 10.1038/s41586-019-1027-4] [Citation(s) in RCA: 432] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 02/26/2019] [Indexed: 11/09/2022]
Abstract
A cure for HIV-1 remains unattainable as only one case has been reported, a decade ago1,2. The individual-who is known as the 'Berlin patient'-underwent two allogeneic haematopoietic stem-cell transplantation (HSCT) procedures using a donor with a homozygous mutation in the HIV coreceptor CCR5 (CCR5Δ32/Δ32) to treat his acute myeloid leukaemia. Total body irradiation was given with each HSCT. Notably, it is unclear which treatment or patient parameters contributed to this case of long-term HIV remission. Here we show that HIV-1 remission may be possible with a less aggressive and toxic approach. An adult infected with HIV-1 underwent allogeneic HSCT for Hodgkin's lymphoma using cells from a CCR5Δ32/Δ32 donor. He experienced mild gut graft-versus-host disease. Antiretroviral therapy was interrupted 16 months after transplantation. HIV-1 remission has been maintained over a further 18 months. Plasma HIV-1 RNA has been undetectable at less than one copy per millilitre along with undetectable HIV-1 DNA in peripheral CD4 T lymphocytes. Quantitative viral outgrowth assays from peripheral CD4 T lymphocytes show no reactivatable virus using a total of 24 million resting CD4 T cells. CCR5-tropic, but not CXCR4-tropic, viruses were identified in HIV-1 DNA from CD4 T cells of the patient before the transplant. CD4 T cells isolated from peripheral blood after transplantation did not express CCR5 and were susceptible only to CXCR4-tropic virus ex vivo. HIV-1 Gag-specific CD4 and CD8 T cell responses were lost after transplantation, whereas cytomegalovirus-specific responses were detectable. Similarly, HIV-1-specific antibodies and avidities fell to levels comparable to those in the Berlin patient following transplantation. Although at 18 months after the interruption of treatment it is premature to conclude that this patient has been cured, these data suggest that a single allogeneic HSCT with homozygous CCR5Δ32 donor cells may be sufficient to achieve HIV-1 remission with reduced intensity conditioning and no irradiation, and the findings provide further support for the development of HIV-1 remission strategies based on preventing CCR5 expression.
Collapse
Affiliation(s)
- Ravindra K Gupta
- Division of Infection and Immunity, UCL, London, UK.
- Department of Infection, UCLH, London, UK.
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Africa Health Research Institute, Durban, South Africa.
| | | | | | - Hoi Ping Mok
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dimitra Peppa
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - 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
| | - Monique Nijhuis
- Translational Virology, Department of Medical Microbiology, University Medical Center, Utrecht, The Netherlands
| | - Annemarie M J Wensing
- Translational Virology, Department of Medical Microbiology, University Medical Center, Utrecht, The Netherlands
| | - Helen Lee
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Paul Grant
- Department of Virology, UCLH, London, UK
| | | | | | - Matthew Pace
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Fanny Salasc
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Andrew J Innes
- Department of Clinical Haematology, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
- Imperial College London, London, UK
| | - Luke Muir
- Division of Infection and Immunity, UCL, London, UK
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, UK
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrew M L Lever
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Simon G Edwards
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, UK
| | - Ian H Gabriel
- Department of Clinical Haematology, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
- Imperial College London, London, UK
- Department of Haematology, Chelsea and Westminster Hospitals Foundation NHS Trust, London, UK
| | - Eduardo Olavarria
- Department of Clinical Haematology, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
- Imperial College London, London, UK
| |
Collapse
|
8
|
Stekler JD, Tapia K, Maenza J, Stevens CE, Ure GA, O'Neal JD, Lane A, Mullins JI, Coombs RW, Holte S, Collier AC. No Time to Delay! Fiebig Stages and Referral in Acute HIV infection: Seattle Primary Infection Program Experience. AIDS Res Hum Retroviruses 2018; 34:657-666. [PMID: 29756456 DOI: 10.1089/aid.2017.0276] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There has been increasing recognition of the importance of diagnosing individuals during the earliest stages of human immunodeficiency virus (HIV) infection. Sera from individuals referred to a primary HIV infection research program were screened using the IgG-sensitive Vironostika HIV-1 Microelisa System, IgG/IgM-sensitive GS HIV-1/HIV-2 Plus O antibody enzyme immunoassay (EIA), or Abbott ARCHITECT HIV antigen (Ag)/antibody (Ab) Combo assay and confirmed by the Bio-Rad Multispot and Western blot. A subset of participants was co-enrolled in a study designed to compare the ability of point-of-care tests to detect early infection. We calculated time within primary infection laboratory stages using actual observed transitions and with an expectation-maximization algorithm. Three hundred and sixty participants contributed data to this analysis. Of 123 persons referred with EIA-negative/RNA-positive test results (Fiebig stage I-II) or for concern for symptoms, 24 (20%) were still in stages I-II, and 99 (80%) were in stages III or later at their screening visit. Participants were estimated to spend a median of 13.5 days in stages I and II, 2.3 days in stage III, and 7.8 days in stage IV. OraQuick performed on oral fluids detected 53% of 17 participants in stage V. The durations of stages we observed are consistent with previous publications. Most persons referred for research no longer had acute infection at their first visit. Programs wishing to identify persons in the very earliest stages of infection need to expedite referrals or develop targeted screening programs.
Collapse
Affiliation(s)
- Joanne D. Stekler
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Epidemiology, University of Washington, Seattle, Washington
- Department of Global Health, University of Washington, Seattle, Washington
| | - Kenneth Tapia
- Department of Global Health, University of Washington, Seattle, Washington
| | - Janine Maenza
- Department of Medicine, University of Washington, Seattle, Washington
| | - Claire E. Stevens
- Department of Medicine, University of Washington, Seattle, Washington
| | - George A. Ure
- Department of Medicine, University of Washington, Seattle, Washington
| | | | - Aric Lane
- Department of Medicine, University of Washington, Seattle, Washington
| | - James I. Mullins
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Microbiology, University of Washington, Seattle, Washington
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Robert W. Coombs
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Sarah Holte
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ann C. Collier
- Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
9
|
Rozera G, Fabbri G, Lorenzini P, Mastrorosa I, Timelli L, Zaccarelli M, Amendola A, Vergori A, Plazzi MM, Cicalini S, Antinori A, Capobianchi MR, Abbate I, Ammassari A. Peripheral blood HIV-1 DNA dynamics in antiretroviral-treated HIV/HCV co-infected patients receiving directly-acting antivirals. PLoS One 2017; 12:e0187095. [PMID: 29077766 PMCID: PMC5659787 DOI: 10.1371/journal.pone.0187095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/15/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Aim was to determine the dynamics of peripheral blood mononuclear cells (PBMC)- associated total HIV-1 DNA in successfully ART-treated HIV/HCV co-infected patients receiving DAA treatment and to explore possible virological hypotheses underlying the phenomenon. METHODS Longitudinal, single-centre study measuring total HIV-1 DNA before the start of DAA, at the end of treatment (EOT), and 3 months after treatment. Univariable and multivariable analyses were used to assess factors associated with HIV-1 DNA increase ≥0.5 Log copies/million PBMC. Episomal 2-LTR forms, residual HIV-1 viremia and proviral DNA quasispecies evolution were also investigated. RESULTS 119 successfully ART-treated HIV/HCV co-infected patients were included. Median baseline HIV-1 DNA was 3.84 Log copies/million PBMC (95%CI 3.49-4.05), and no significant variation with respect to baseline was found at EOT and after 3 months of DAA termination. In 17% of cases an increase ≥0.5 Log copies/million PBMC was observed at EOT compared to baseline. HIV-1 DNA increase was independently associated with lower baseline HIV-1 DNA, longer HIV suppression, raltegravir-based ART and previous exposure to interferon/ribavirin for HCV treatment. In none of the patients with HIV-1 DNA increase, 2-LTR forms were detected at baseline, while in 2 cases 2-LTR forms were found at EOT, without association with residual HIV-1 RNA viremia. No evidence of viral evolution was observed. CONCLUSIONS In successfully ART-treated HIV/HCV co-infected patients receiving DAA, PBMC-associated total HIV-1 DNA was quite stable over time, but some patients showed a considerable increase at EOT when compared to baseline. A significantly higher risk of HIV DNA increase was found, in presence of lower cellular HIV reservoir at baseline. Activation of replicative-competent virus generating new rounds of viral replication seems unlikely, while mobilization of cell-associated HIV from tissue reservoirs could be hypothesized.
Collapse
Affiliation(s)
- Gabriella Rozera
- Laboratory of Virology, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Gabriele Fabbri
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Patrizia Lorenzini
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Ilaria Mastrorosa
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Laura Timelli
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Mauro Zaccarelli
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Alessandra Amendola
- Laboratory of Virology, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Alessandra Vergori
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Maria Maddalena Plazzi
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Stefania Cicalini
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Andrea Antinori
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Isabella Abbate
- Laboratory of Virology, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| | - Adriana Ammassari
- Clinical Department, National Institute for Infectious Diseases “L. Spallanzani” IRCCS, Rome, Italy
| |
Collapse
|
10
|
Rossetti B, Meini G, Bianco C, Lamonica S, Mondi A, Belmonti S, Fanti I, Ciccarelli N, Di Giambenedetto S, Zazzi M, De Luca A. Total cellular HIV-1 DNA decreases after switching to raltegravir-based regimens in patients with suppressed HIV-1 RNA. J Clin Virol 2017; 91:18-24. [PMID: 28395180 DOI: 10.1016/j.jcv.2017.03.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/08/2017] [Accepted: 03/20/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND The integrase inhibitor raltegravir has been used to intensify antiretroviral therapy in patients with undetectable plasma HIV-1RNA, resulting in variable perturbation of HIV-1 nucleic acids levels in peripheral blood. OBJECTIVES We aimed at monitoring residual plasma HIV-1RNA and total cellular HIV-1DNA in virologically suppressed patients switching to raltegravir-based regimens. STUDY DESIGN Fifty-eight subjects on protease inhibitor (PI) or nonnucleoside reverse transcriptase inhibitor (NNRTI)-based regimens, with plasma HIV-1RNA levels <40 copies/ml for ≥6 months and CD4 counts >200cells/μl for ≥12 months were enrolled. Thirty-four patients were from the treatment simplification RASTA randomized study switching standard therapy to a raltegravir-based regimen (RASTA group), while 24 continued a PI or NNRTI based-regimen (controls). Residual plasma HIV-1RNA (5-40copies/mL) and HIV-1DNA were assessed at 0, 24 and 48 weeks. RESULTS At week 0 (W0), HIV-1DNA was detected in all patients while at W48 it was detectable in 82.4% of the RASTA group vs 100% of controls (p=0.03). There was a significant decline of HIV-1DNA at W48 in the RASTA group (mean change from baseline -0.21 [95% CI -0.41; -0.01] log10 copies/106 CD4; p=0.03) but not in controls. Ultrasensitive HIV-1RNA was detectable at baseline in 50% of RASTA group vs 67% of controls and at W48 in 32.4% vs 42%, respectively. No differences were found between HIV-1RNA levels at baseline and W48 within and between groups. CONCLUSIONS Switching successful therapy to raltegravir-based regimens may be associated with a decrease of the HIV-1 reservoir, as measured by peripheral blood cellular HIV-1DNA levels.
Collapse
Affiliation(s)
- Barbara Rossetti
- Infectious Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy; Clinic of Infectious Diseases, Catholic University of Sacred Heart, Rome, Italy.
| | - Genny Meini
- Medical Biotechnology Department, University of Siena, Siena, Italy
| | - Claudia Bianco
- Infectious Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Silvia Lamonica
- Clinic of Infectious Diseases, Catholic University of Sacred Heart, Rome, Italy
| | - Annalisa Mondi
- Clinic of Infectious Diseases, Catholic University of Sacred Heart, Rome, Italy
| | - Simone Belmonti
- Clinic of Infectious Diseases, Catholic University of Sacred Heart, Rome, Italy
| | - Iuri Fanti
- Clinic of Infectious Diseases, Catholic University of Sacred Heart, Rome, Italy
| | | | | | - Maurizio Zazzi
- Medical Biotechnology Department, University of Siena, Siena, Italy
| | - Andrea De Luca
- Infectious Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy; Clinic of Infectious Diseases, Catholic University of Sacred Heart, Rome, Italy; Medical Biotechnology Department, University of Siena, Siena, Italy
| |
Collapse
|
11
|
Sauné K, Raymond S, Boineau J, Pasquier C, Izopet J. Detection and quantification of HIV-1 RNA with a fully automated transcription-mediated-amplification assay. J Clin Virol 2016; 84:70-73. [PMID: 27728849 DOI: 10.1016/j.jcv.2016.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND Nucleic acid testing is the major method used to monitor HIV viral load. Commercial systems based on real-time PCR assays are available for high-volume centralized laboratory testing, but they are not fully automated. OBJECTIVES AND STUDY DESIGN We have compared the diagnostic performance of the Hologic Aptima HIV-1 Quant Dx assay (Aptima) (based on real-time TMA) on the Panther instrument, a fully-automated random access platform, to that of, the Roche Cobas Ampliprep Cobas TaqMan (CAP/CTM) HIV-1 version 2.0 (based on real-time PCR). RESULTS Probit analysis of replicate dilutions of NIBSC WHO International HIV-1 Standard, gave LODs of 8.6 c/ml for Aptima and 15.2 c/ml for CAP/CTM. The agreement between the assays was excellent when measuring HIV RNA in a calibrated reference (κ=0.90, p<0.001) and good when measuring clinical samples (κ=0.62, p<0.001). The correlation among the samples quantified by the two methods was very good (r=0.95, p<0.001) and the mean difference between the values obtained with the two assays was 0.02 log c/ml for B and non-B subtypes. The vast majority of results showed <0.5 log variance between the two assays (89%); only one sample showed results that differed by over 1.0 log c/ml. CONCLUSION The performance of the new fully automated Aptima assay is adequate for clinical monitoring of HIV-1 RNA during infections and treatment. The Aptima assay is well suited for routine laboratory use.
Collapse
Affiliation(s)
- K Sauné
- INSERM, U1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France; CHU Toulouse, Hôpital Purpan, Laboratoire de Virologie, Institut Fédératif de Biologie, Toulouse, France.
| | - S Raymond
- INSERM, U1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France; CHU Toulouse, Hôpital Purpan, Laboratoire de Virologie, Institut Fédératif de Biologie, Toulouse, France
| | - J Boineau
- CHU Toulouse, Hôpital Purpan, Laboratoire de Virologie, Institut Fédératif de Biologie, Toulouse, France
| | - C Pasquier
- INSERM, U1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France; CHU Toulouse, Hôpital Purpan, Laboratoire de Virologie, Institut Fédératif de Biologie, Toulouse, France
| | - J Izopet
- INSERM, U1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France; CHU Toulouse, Hôpital Purpan, Laboratoire de Virologie, Institut Fédératif de Biologie, Toulouse, France
| |
Collapse
|
12
|
Dustin LB, Bartolini B, Capobianchi MR, Pistello M. Hepatitis C virus: life cycle in cells, infection and host response, and analysis of molecular markers influencing the outcome of infection and response to therapy. Clin Microbiol Infect 2016; 22:826-832. [PMID: 27592089 PMCID: PMC5627509 DOI: 10.1016/j.cmi.2016.08.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 08/16/2016] [Accepted: 08/25/2016] [Indexed: 12/13/2022]
Abstract
Hepatitis C virus (HCV) is a major global health burden accounting for around 170 million chronic infections worldwide. Since its discovery, which dates back to about 30 years ago, many details of the viral genome organization and the astonishing genetic diversity have been unveiled but, owing to the difficulty of culturing HCV in vitro and obtaining fully susceptible yet immunocompetent in vivo models, we are still a long way from the full comprehension of viral life cycle, host cell pathways facilitating or counteracting infection, pathogenetic mechanisms in vivo, and host defences. Here, we illustrate the viral life cycle into cells, describe the interplay between immune and genetic host factors shaping the course of infection, and provide details of the molecular approaches currently used to genotype, monitor replication in vivo, and study the emergence of drug-resistant viral variants.
Collapse
Affiliation(s)
- L B Dustin
- Kennedy Institute for Rheumatology and Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - B Bartolini
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - M R Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - M Pistello
- Virology Unit, Pisa University Hospital, and Virology Section and Retrovirus Centre, Department of Translational Research, University of Pisa, Pisa, Italy.
| |
Collapse
|
13
|
Amendola A, Pisciotta M, Aleo L, Ferraioli V, Angeletti C, Capobianchi MR. Evaluation of the Aptima(®) HIV-1 Quant Dx assay for HIV-1 RNA viral load detection and quantitation in plasma of HIV-1-infected individuals: A comparison with Abbott RealTime HIV-1 assay. J Med Virol 2016; 88:1535-44. [PMID: 26864171 PMCID: PMC6585778 DOI: 10.1002/jmv.24493] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2016] [Indexed: 11/16/2022]
Abstract
The Hologic Aptima® HIV‐1 Quant Dx assay (Aptima HIV) is a real‐time transcription‐mediated amplification method CE‐approved for use in diagnosis and monitoring of HIV‐1 infection. The analytical performance of this new assay was compared to the FDA‐approved Abbott RealTime HIV‐1 (RealTime). The evaluation was performed using 220 clinical plasma samples, the WHO 3rd HIV‐1 International Standard, and the QCMD HIV‐1 RNA EQA. Concordance on qualitative results, correlation between quantitative results, accuracy, and reproducibility of viral load data were analyzed. The ability to measure HIV‐1 subtypes was assessed on the second WHO International Reference Preparation Panel for HIV‐1 Subtypes. With clinical samples, inter‐assay agreement for qualitative results was high (91.8%) with Cohen's kappa statistic equal to 0.836. For samples with quantitative results in both assays (n = 93), Lin's concordance correlation coefficient was 0.980 (P
< 0.0001) and mean differences of measurement, conducted according to Bland–Altman method, was low (0.115 log10 copies/ml). The Aptima HIV quantified the WHO 3rd HIV‐1 International Standard diluted from 2000 to 31 cp/ml (5,700–88 IU/ml) at expected values with excellent linearity (R2 > 0.970) and showed higher sensitivity compared to RealTime being able to detect HIV‐1 RNA in 10 out of 10 replicates containing down to 7 cp/ml (20 IU/ml). Reproducibility was very high, even at low HIV‐1 RNA values. The Aptima HIV was able to detect and accurately quantify all the main HIV‐1 subtypes in both reference panels and clinical samples. Besides excellent performance, Aptima HIV shows full automation, ease of use, and improved workflow compared to RealTime. J. Med. Virol. 88:1535–1544, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alessandra Amendola
- Laboratory of VirologyNational Institute for Infectious Diseases “Lazzaro Spallanzani,”RomeItaly
| | - Maria Pisciotta
- Laboratory of VirologyNational Institute for Infectious Diseases “Lazzaro Spallanzani,”RomeItaly
| | - Loredana Aleo
- Laboratory of VirologyNational Institute for Infectious Diseases “Lazzaro Spallanzani,”RomeItaly
| | - Valeria Ferraioli
- Laboratory of VirologyNational Institute for Infectious Diseases “Lazzaro Spallanzani,”RomeItaly
| | - Claudio Angeletti
- Department of EpidemiologyNational Institute for Infectious Diseases “Lazzaro Spallanzani,”RomeItaly
| | | |
Collapse
|
14
|
Amendola A, Marsella P, Bloisi M, Forbici F, Angeletti C, Capobianchi MR. Ability of two commercially available assays (Abbott RealTime HIV-1 and Roche Cobas AmpliPrep/Cobas TaqMan HIV-1 Version 2.0) to quantify low HIV-1 RNA Levels (<1,000 copies/milliliter): comparison with clinical samples and NIBSC working reagent for nucleic acid testing assays. J Clin Microbiol 2014; 52:2019-26. [PMID: 24671791 PMCID: PMC4042785 DOI: 10.1128/jcm.00288-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/21/2014] [Indexed: 11/20/2022] Open
Abstract
Concordance between molecular assays may be suboptimal at low HIV-1 viremia levels (<1,000 copies/ml); therefore, it may be difficult to define and compare virologic endpoints for successful and failed therapy. We compared two commercial assays (the Abbott RealTime HIV-1 and the Roche Cobas AmpliPrep/TaqMan HIV-1 version 2.0) for their ability to detect and quantify low viral loads. A comparison was performed using 167 residual clinical samples (with values ranging from "not detected" to 1,000 copies/ml, as measured by the Abbott assay) and the National Institute and Biological Standards and Control (NIBSC) HIV-1 RNA working reagent 1 for nucleic acid amplification techniques (NAT) assays (serially diluted to a range from 1 to 1,000 copies/ml). Quantitative results were compared using Lin's concordance correlation coefficient and a Bland-Altman plot. Concordance with the qualitative results was measured by Cohen's kappa statistic. With clinical samples, the degree of interassay concordance of the qualitative results at a 40-copies/ml HIV-1 RNA threshold was substantial (κ = 0.762); the correlation among the quantified samples was suboptimal (concordance correlation coefficient, 0.728; P < 0.0001); the mean difference of the values between the Roche and Abbott assays was 0.193 log10 copies/ml. Using the HIV-1 RNA working reagent 1 for NAT assays, the results provided by the Roche assay were, on average, 3 times higher than expected, while the Abbott assay showed high accuracy. The Roche assay was highly sensitive, being able to detect a level as low as 3.5 copies/ml HIV-1 RNA with 95% probability. The performance characteristics of each molecular assay should be taken into account when HIV-1 RNA threshold values for "virologic suppression," "virologic failure," "persistent low viral loads," etc., are defined and indicated in the support of clinical decisions.
Collapse
Affiliation(s)
| | - Patrizia Marsella
- National Institute for Infectious Diseases Lazzaro Spallanzani, Rome, Italy
| | - Maria Bloisi
- National Institute for Infectious Diseases Lazzaro Spallanzani, Rome, Italy
| | - Federica Forbici
- National Institute for Infectious Diseases Lazzaro Spallanzani, Rome, Italy
| | - Claudio Angeletti
- National Institute for Infectious Diseases Lazzaro Spallanzani, Rome, Italy
| | | |
Collapse
|
15
|
Swenson LC, Cobb B, Geretti AM, Harrigan PR, Poljak M, Seguin-Devaux C, Verhofstede C, Wirden M, Amendola A, Boni J, Bourlet T, Huder JB, Karasi JC, Zidovec Lepej S, Lunar MM, Mukabayire O, Schuurman R, Tomažič J, Van Laethem K, Vandekerckhove L, Wensing AMJ. Comparative performances of HIV-1 RNA load assays at low viral load levels: results of an international collaboration. J Clin Microbiol 2014; 52:517-23. [PMID: 24478482 PMCID: PMC3911321 DOI: 10.1128/jcm.02461-13] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 11/26/2013] [Indexed: 11/20/2022] Open
Abstract
Low-level viremia during antiretroviral therapy and its accurate measurement are increasingly relevant. Here, we present an international collaboration of 4,221 paired blood plasma viral load (pVL) results from four commercial assays, emphasizing the data with low pVL. The assays compared were the Abbott RealTime assay, the Roche Amplicor assay, and the Roche TaqMan version 1 and version 2 assays. The correlation between the assays was 0.90 to 0.97. However, at a low pVL, the correlation fell to 0.45 to 0.85. The observed interassay concordance was higher when detectability was defined as 200 copies/ml than when it was defined as 50 copies/ml. A pVL of ∼100 to 125 copies/ml by the TaqMan version 1 and version 2 assays corresponded best to a 50-copies/ml threshold with the Amplicor assay. Correlation and concordance between the viral load assays were lower at a low pVL. Clear guidelines are needed on the clinical significance of low-level viremia.
Collapse
Affiliation(s)
- Luke C. Swenson
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Bryan Cobb
- Roche Molecular Systems, Pleasanton, California, USA
| | - Anna Maria Geretti
- Institute of Infection and Global Health, University of Liverpool, United Kingdom
| | | | - Mario Poljak
- University of Ljubljana, Institute of Microbiology and Immunology, Ljubljana, Slovenia
| | | | | | - Marc Wirden
- AP-HP, Pitié-Salpêtrière Hospital, INSERM U 943, and Pierre et Marie Curie University Paris, Paris, France
| | - Alessandra Amendola
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani, Rome, Italy
| | - Jurg Boni
- Institute of Medical Virology, Swiss National Center for Retroviruses, University of Zurich, Zurich, Switzerland
| | - Thomas Bourlet
- Laboratory of Virology, CHU de Saint-Etienne, Saint-Etienne, France
| | - Jon B. Huder
- Institute of Medical Virology, Swiss National Center for Retroviruses, University of Zurich, Zurich, Switzerland
| | | | | | - Maja M. Lunar
- University of Ljubljana, Institute of Microbiology and Immunology, Ljubljana, Slovenia
| | | | - Rob Schuurman
- University Medical Center Utrecht, Utrecht, Netherlands
| | - Janez Tomažič
- University of Ljubljana, Institute of Microbiology and Immunology, Ljubljana, Slovenia
| | - Kristel Van Laethem
- Clinical and Epidemiological Virology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
16
|
Quantification of viral loads lower than 50 copies per milliliter by use of the Cobas AmpliPrep/Cobas TaqMan HIV-1 test, version 2.0, can predict the likelihood of subsequent virological rebound to >50 copies per milliliter. J Clin Microbiol 2013; 51:1555-7. [PMID: 23390288 DOI: 10.1128/jcm.00100-13] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
After 1 year of follow-up, patients on HAART with a baseline viral load (VL) of <20 copies/ml showed significantly lower odds of virological rebound to two consecutive VLs of >50 copies/ml than those with baseline VLs of 20 to 39 and 40 to 49 (P < 0.001). The time to virological rebound was also significantly shorter (P < 0.001) for the groups with baseline VLs of 20 to 39 and 40 to 49.
Collapse
|
17
|
Ruelle J, Debaisieux L, Vancutsem E, De Bel A, Delforge ML, Piérard D, Goubau P. HIV-1 low-level viraemia assessed with 3 commercial real-time PCR assays show high variability. BMC Infect Dis 2012; 12:100. [PMID: 22530816 PMCID: PMC3445837 DOI: 10.1186/1471-2334-12-100] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 04/24/2012] [Indexed: 01/29/2023] Open
Abstract
Background Current real-time PCR-based HIV-1 viral load (VL) assays allow the detection of residual viraemia in antiretroviral-treated patients. The clinical outcome of HIV1 patients experiencing low-level replication (<50 cop/mL) in comparison with fully suppressed patients is currently debated. We analysed variability of 3 VL assays <50 cop/mL, and evaluated the reproducibility of viral blips <100 cop/mL. Methods Three commercial VL assays were tested: Versant HIV-1 RNA 1.0 kPCR (Siemens), Abbott Realtime HIV-1, and Cobas Ampliprep/Cobas Taqman HIV-1 v2.0 (Roche). Ten replicates of a reference sample at 4 low target dilutions were tested to evaluate assay variability. Prospective collection of 181 clinical samples with detectable VL <50 cop/mL was used to evaluate intra-and inter-assay variability by triplicate testing. Samples from 26 patients experiencing a viral blip were retested. Results All assays showed substantial variability at low VL level: the coefficient of variation at 100, 50, 25 and 12 cop/mL ranged respectively from 32 to 44%, 35 to 68%, 41 to 83% and 33 to 77%. In the intra-assay evaluation of repeatability, 52.5 to 57.5% of detectable VL <50 cop/mL tested in triplicate showed at least one fully undetected result. Variability was similar in the inter-assay arm. The VL blips could only be reproduced in 19% of cases. Conclusions The most recent versions of widespread commercial VL assays showed substantial variability at low levels and residual viraemia could not be consistently reproduced. Patient outcome studies comparing residual VL to full suppression are therefore biased when using commercial assays.
Collapse
Affiliation(s)
- Jean Ruelle
- UCLouvain, AIDS Reference Laboratory, Avenue Hippocrate 54 - B1.54.05, 1200 Brussels, Belgium.
| | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The source, significance and optimal management of low-level viraemia during highly active antiretroviral therapy (HAART) are poorly defined. This review highlights recent observations that have implications for clinical practice. RECENT FINDINGS The definition of low-level viraemia remains elusive. Whereas evidence obtained with second-generation viral load assays indicates that confirmed detection of plasma HIV-1 RNA above 50 copies/ml predicts negative outcomes, third-generation assays detect HIV-1 RNA below this threshold. In patients monitored with the Abbott RealTime assay, the cutoff that should trigger confirmation of viraemia and clinical review can be revised to 40 copies/ml. Further data are needed on the cost-effectiveness of intervening when RNA detection is observed below this cutoff. Discrepancies among viral load assays prevent generalization of these observations. To further compound the issue, most patients on stably suppressive HAART show residual viraemia at around 1-10 copies/ml using research-based ultrasensitive assays. The source of residual viraemia remains controversial, but neither short nor long-term HAART intensification with antiretrovirals such as raltegravir reduces the viraemia. A transient effect of intravenous immunoglobulin has been reported, and different regimens may vary in their propensity to allow HIV-1 RNA persistence. Further studies are required to clarify the relationship between low-level viraemia and the size of proviral DNA reservoirs, and the contribution of cellular and tissue compartments and cell-to-cell spread to ongoing virus replication during HAART. SUMMARY Understanding the source and clinical significance of HIV-1 RNA persistence in plasma during HAART is required to guide patient care and inform the design of HIV eradication strategies.
Collapse
|