1
|
Lapointe HR, Mwimanzi F, Cheung PK, Sang Y, Yaseen F, Umviligihozo G, Kalikawe R, Speckmaier S, Moran-Garcia N, Datwani S, Duncan MC, Agafitei O, Ennis S, Young L, Ali H, Ganase B, Omondi FH, Dong W, Toy J, Sereda P, Burns L, Costiniuk CT, Cooper C, Anis AH, Leung V, Holmes DT, DeMarco ML, Simons J, Hedgcock M, Prystajecky N, Lowe CF, Pantophlet R, Romney MG, Barrios R, Guillemi S, Brumme CJ, Montaner JSG, Hull M, Harris M, Niikura M, Brockman MA, Brumme ZL. People With Human Immunodeficiency Virus Receiving Suppressive Antiretroviral Therapy Show Typical Antibody Durability After Dual Coronavirus Disease 2019 Vaccination and Strong Third Dose Responses. J Infect Dis 2023; 227:838-849. [PMID: 35668700 PMCID: PMC9214159 DOI: 10.1093/infdis/jiac229] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/19/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Longer-term humoral responses to 2-dose coronavirus disease 2019 (COVID-19) vaccines remain incompletely characterized in people living with human immunodeficiency virus (HIV) (PLWH), as do initial responses to a third dose. METHODS We measured antibodies against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein receptor-binding domain, angiotensin-converting enzyme 2 (ACE2) displacement, and viral neutralization against wild-type and Omicron strains up to 6 months after 2-dose vaccination, and 1 month after the third dose, in 99 PLWH receiving suppressive antiretroviral therapy and 152 controls. RESULTS Although humoral responses naturally decline after 2-dose vaccination, we found no evidence of lower antibody concentrations or faster rates of antibody decline in PLWH compared with controls after accounting for sociodemographic, health, and vaccine-related factors. We also found no evidence of poorer viral neutralization in PLWH after 2 doses, nor evidence that a low nadir CD4+ T-cell count compromised responses. Post-third-dose humoral responses substantially exceeded post-second-dose levels, though Omicron-specific responses were consistently weaker than responses against wild-type virus. Nevertheless, post-third-dose responses in PLWH were comparable to or higher than controls. An mRNA-1273 third dose was the strongest consistent correlate of higher post-third-dose responses. CONCLUSION PLWH receiving suppressive antiretroviral therapy mount strong antibody responses after 2- and 3-dose COVID-19 vaccination. Results underscore the immune benefits of third doses in light of Omicron.
Collapse
Affiliation(s)
- Hope R Lapointe
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Francis Mwimanzi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Peter K Cheung
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Yurou Sang
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Fatima Yaseen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Gisele Umviligihozo
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Rebecca Kalikawe
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Sarah Speckmaier
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Nadia Moran-Garcia
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Sneha Datwani
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Maggie C Duncan
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Olga Agafitei
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Siobhan Ennis
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Landon Young
- Division of Medical Microbiology and Virology, St Paul’s Hospital, Vancouver, British Columbia, Canada
| | - Hesham Ali
- John Ruedy Clinic, St Paul’s Hospital, Vancouver, British Columbia, Canada
| | - Bruce Ganase
- AIDS Research Program, St Paul’s Hospital, Vancouver, British Columbia, Canada
| | - F Harrison Omondi
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Winnie Dong
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Junine Toy
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Paul Sereda
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Laura Burns
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, British Columbia, Canada
| | - Cecilia T Costiniuk
- Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Curtis Cooper
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Aslam H Anis
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
- CIHR Canadian HIV Trials Network, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Health Evaluation and Outcome Sciences, Vancouver, British Columbia, Canada
| | - Victor Leung
- Division of Medical Microbiology and Virology, St Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel T Holmes
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mari L DeMarco
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Janet Simons
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Natalie Prystajecky
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Vancouver, British Columbia, Canada
| | - Christopher F Lowe
- Division of Medical Microbiology and Virology, St Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ralph Pantophlet
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Marc G Romney
- Division of Medical Microbiology and Virology, St Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rolando Barrios
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Silvia Guillemi
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Department of Family Practice, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chanson J Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julio S G Montaner
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark Hull
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Department of Family Practice, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mark A Brockman
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Zabrina L Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
2
|
Kouamou V, Gundidza P, Ndhlovu CE, Makadzange AT, Study C. Effects of Gender and Baseline CD4 Count on Post Treatment CD4 Count Recovery and Outcomes in Patients with Advanced HIV Disease: a Retrospective Cohort Study. AIDS Res Hum Retroviruses 2023. [PMID: 36924288 DOI: 10.1089/aid.2022.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Presentation to care with advanced HIV disease (AHD) is a significant problem in Sub-Saharan Africa. We evaluated factors associated with immune recovery among individuals presenting to care with AHD in Zimbabwe. We conducted a retrospective evaluation of outcomes among adult (>18 years old) individuals with AHD (CD4 count ≤ 200 cells/mm3) receiving care at 18 outpatient primary care clinics in Harare, Zimbabwe. Baseline and 12-month CD4 count data were extracted from medical records. CD4 count recovery (defined as CD4 count > 200 cells/mm3) after 12 months on NNRTI-based antiretroviral therapy (ART) regimen was determined and factors associated with CD4 count recovery were established using logistic regression. All statistical analysis was performed on SPSS v23. A total of 1338 participant records were included in the analysis. The median interquartile range (IQR) age was 37 (30-43) years and 52% were females. The baseline median (IQR) CD4 count was 50 (28-75) cells/mm3 and was significantly lower among patients with history of cryptococcal meningitis compared to those without (25(10-52) vs 52(32-77), respectively; p=0.0009). The median (IQR) CD4 count at 12 months after ART initiation increased from 50 (28-75) at baseline to 180 (92-290) cells/mm3. Immune recovery with a CD4 count > 200 cells/mm3, was observed in 181/417 (43%). Male gender and low baseline CD4 count were strong predictors of poor immunological recovery on ART. Immunological recovery following ART initiation was 43% among individuals with AHD. Male patients are most vulnerable to persistent immunological failure.
Collapse
Affiliation(s)
- Vinie Kouamou
- University of Zimbabwe Faculty of Medicine, 108329, Primary Health Care Sciences, Harare, Harare, Zimbabwe.,Charles River Medical Group, Harare, Zimbabwe;
| | | | - Chiratidzo Ellen Ndhlovu
- Charles River Medical Group, Harare, -, Zimbabwe.,University of Zimbabwe Faculty of Medicine, 108329, Harare, Harare, Zimbabwe;
| | | | | |
Collapse
|
3
|
Yuan R, Li L, Hu W, Zhuang K, Zhang E, Yan Y, Feng L, Chen X, Cao Q, Ke H, Gui X, Yang R. Characteristics of refined lymphocyte subsets changes in people living with HIV/AIDS during antiretroviral therapy period: An observation from Wuhan, China. Front Immunol 2023; 14:1089379. [PMID: 36845097 PMCID: PMC9948076 DOI: 10.3389/fimmu.2023.1089379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Background To analyze the changing characteristics of continuous monitoring of refined lymphocyte subsets in people living with HIV/AIDS (PLWHA) during ART period. Methods Refined lymphocyte subsets was continuously monitored using flow cytometry for 173 PLWHA, who were hospitalized in Zhongnan Hospital of Wuhan University from August 17, 2021 to September 14, 2022. The effect of ART status and duration of ART on changes of refined lymphocyte subsets were compared in different groups. Then, the levels of refined lymphocyte subsets in PLWHA treated for more than 10 years were compared to those of 1086 healthy individuals. Results In addition to conventional CD4+ T lymphocytes and CD4+/CD8+ ratio, gradually increasing in numbers of CD3+CD4+CD45RO cells, CD3+CD4+CD45RA cells, CD45RA+CD3+CD4+CD25+CD127low and CD45RO+CD3+CD4+CD25+CD127low cells were found with the increase of ART duration. The number of CD4+CD28+ cells and CD8+CD28+ cells were 174/ul and 233/ul at 6 months post-ART, which gradually increased to 616/ul and 461/ul after ART initiation more than 10 years. Moreover, in ART ≤ 6 months, 6 months-3years, 3-10 years and >10 years groups, the percentage of CD3+CD8+HLA-DR+/CD8 were 79.66%, 69.73%, 60.19% and 57.90%, respectively, and the differences between groups showed statistical significance (F=5.727, P=0.001). For those PLWHA with ART more than 10 years, the levels of CD4+ T lymphocytes, CD3+CD4+CD45RO cells, CD3+CD4+CD45RA cells, CD4+CD28+ cells and CD8+CD28+ cells can increase to levels similar to those of healthy control. However, for those PLWHA with ART more than 10 years, CD4+/CD8+ ratio was 0.86 ± 0.47, which was lower than that of healthy control (0.86 ± 0.47 vs 1.32 ± 0.59, t=3.611, P=0.003); absolute counts and percentage of CD3+CD8+HLA-DR+ cells were 547/ul and 57.90%, which were higher than those of healthy control(547/ul vs 135/ul, t=3.612, P=0.003; 57.90% vs 22.38%, t=6.959, P<0.001). Conclusion Persistent ART can gradually improve the immune status of PLWHA, which is manifested in the increase of lymphocytes, function recovery of lymphocytes and reduction of aberrant activation status of the immune system. After 10 years of standardized ART, most lymphocytes could return to levels of healthy persons, although it may take longer to complete recovery for CD4+/CD8+ ratio and CD3+CD8+HLA-DR+ cells.
Collapse
Affiliation(s)
- Rui Yuan
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ling Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenjia Hu
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ke Zhuang
- Animal Biosafety Shelter Laboratory (ABSL)‐III Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Ejuan Zhang
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yajun Yan
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ling Feng
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoping Chen
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Cao
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hengning Ke
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xien Gui
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rongrong Yang
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China,*Correspondence: Rongrong Yang,
| |
Collapse
|
4
|
Lu L, Li X, Liu X, Qiu Z, Han Y, Song X, Li Y, Li X, Cao W, Lv W, Dou Z, Li T. The pattern and magnitude of T cell subsets reconstitution during ten years of ART with viral suppression in HIV-infected patients. Aging (Albany NY) 2022; 14:9647-9667. [PMID: 36490352 PMCID: PMC9792206 DOI: 10.18632/aging.204416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND The extent of immune reconstitution in human immunodeficiency virus (HIV) infected persons receiving long-term antiretroviral therapy (ART) with controlled viral load has been controversial. We studied the extent and speed of T cell subsets retrieval after long-term antiretroviral treatment. METHODS 662 HIV-infected patients followed at least 2 years whose plasma HIV-1 RNA load <50 copies/mL were evaluated for longitudinal and functional phenotypic indices of immune restoration. Determinants of change in magnitude and importance of recovery have been evaluated using mixed linear regression models. RESULTS Almost all robust immune restorations achieved occurred after 2-3 years of ART. The median CD4 lymphocyte count increased 449 cells/μl (IQR 303-604) from 226 cells/μl (IQR 83-336) at baseline during the third year (P < 0.001); CD4+T lymphocyte rises during the sixth and tenth years were not significant. Naive and memory CD4+T cells'reconstitution occurred in the sixth and eighth years of ART but no significant change thereafter. The change of CD45RA+Naïve and CD45RA-memory CD4+T cell reconstitution is different in baseline CD4+T cell counts <100 cells/μl group and in baseline CD4+T cell counts >100 cells/μl group. Activation antigen expression (CD38 or HLA-DR) on CD8 lymphocytes declined mostly during the first till second year, and after 4 years, activation antigen expression on patient lymphocytes showed no significant change. The proportion of CD4 cells expressing CD28 climbed during the first years and reached normal levels in the second year. CONCLUSIONS Immune restoration was dependent on the capacity of immune system during the first 2-3 year of ART. But the significant change of CD4 and compartments of CD4+T cells could persist until 6-8 years. The pattern of CD38+CD8+, HLA-DR+CD8+, CD28+CD4+ T cells could quickly return to normal level and no significant change after sufficient time of ART. In general, the immune response compared to the baseline status may be the overall effect from the age and time of antiretroviral treatment.
Collapse
Affiliation(s)
- Lianfeng Lu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaodi Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosheng Liu
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhifeng Qiu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Han
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojing Song
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanling Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxia Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Lv
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihui Dou
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China,Tsinghua-Peking Center for Life Sciences, Beijing, China,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Vergori A, Cozzi Lepri A, Cicalini S, Matusali G, Bordoni V, Lanini S, Meschi S, Iannazzo R, Mazzotta V, Colavita F, Mastrorosa I, Cimini E, Mariotti D, De Pascale L, Marani A, Gallì P, Garbuglia A, Castilletti C, Puro V, Agrati C, Girardi E, Vaia F, Antinori A. Immunogenicity to COVID-19 mRNA vaccine third dose in people living with HIV. Nat Commun 2022; 13:4922. [PMID: 35995780 PMCID: PMC9395398 DOI: 10.1038/s41467-022-32263-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/22/2022] [Indexed: 11/09/2022] Open
Abstract
In order to investigate safety and immunogenicity of SARS-CoV-2 vaccine third dose in people living with HIV (PLWH), we analyze anti-RBD, microneutralization assay and IFN-γ production in 216 PLWH on ART with advanced disease (CD4 count <200 cell/mm3 and/or previous AIDS) receiving the third dose of a mRNA vaccine (BNT162b2 or mRNA-1273) after a median of 142 days from the second dose. Median age is 54 years, median CD4 nadir 45 cell/mm3 (20-122), 93% HIV-RNA < 50 c/mL. In 68% of PLWH at least one side-effect, generally mild, is recorded. Humoral response after the third dose was strong and higher than that achieved with the second dose (>2 log2 difference), especially when a heterologous combination with mRNA-1273 as third shot is used. In contrast, cell-mediated immunity remain stable. Our data support usefulness of third dose in PLWH currently receiving suppressive ART who presented with severe immune dysregulation.
Collapse
Affiliation(s)
- Alessandra Vergori
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy.
| | - Alessandro Cozzi Lepri
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation (CREME), Institute for Global Health, UCL, London, UK
| | - Stefania Cicalini
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Veronica Bordoni
- Laboratory of Cellular Immunology and Clinical Pharmacology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Simone Lanini
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Roberta Iannazzo
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Valentina Mazzotta
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Francesca Colavita
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Ilaria Mastrorosa
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Eleonora Cimini
- Laboratory of Cellular Immunology and Clinical Pharmacology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Davide Mariotti
- Laboratory of Cellular Immunology and Clinical Pharmacology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Lydia De Pascale
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Alessandra Marani
- Health Direction, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Paola Gallì
- Health Direction, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - AnnaRosa Garbuglia
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Vincenzo Puro
- Risk Management Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Chiara Agrati
- Laboratory of Cellular Immunology and Clinical Pharmacology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Enrico Girardi
- Scientific Direction, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Francesco Vaia
- Health Direction, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Andrea Antinori
- HIV/AIDS Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
6
|
Lapointe HR, Mwimanzi F, Cheung PK, Sang Y, Yaseen F, Umviligihozo G, Kalikawe R, Speckmaier S, Moran-Garcia N, Datwani S, Duncan MC, Agafitei O, Ennis S, Young L, Ali H, Ganase B, Omondi FH, Dong W, Toy J, Sereda P, Burns L, Costiniuk CT, Cooper C, Anis AH, Leung V, Holmes D, DeMarco ML, Simons J, Hedgcock M, Prystajecky N, Lowe CF, Pantophlet R, Romney MG, Barrios R, Guillemi S, Brumme CJ, Montaner JSG, Hull M, Harris M, Niikura M, Brockman MA, Brumme ZL. People with HIV receiving suppressive antiretroviral therapy show typical antibody durability after dual COVID-19 vaccination, and strong third dose responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022. [PMID: 35350205 PMCID: PMC8963693 DOI: 10.1101/2022.03.22.22272793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background: Longer-term humoral responses to two-dose COVID-19 vaccines remain incompletely characterized in people living with HIV (PLWH), as do initial responses to a third dose. Methods: We measured antibodies against the SARS-CoV-2 spike protein receptor-binding domain, ACE2 displacement and viral neutralization against wild-type and Omicron strains up to six months following two-dose vaccination, and one month following the third dose, in 99 PLWH receiving suppressive antiretroviral therapy, and 152 controls. Results: Though humoral responses naturally decline following two-dose vaccination, we found no evidence of lower antibody concentrations nor faster rates of antibody decline in PLWH compared to controls after accounting for sociodemographic, health and vaccine-related factors. We also found no evidence of poorer viral neutralization in PLWH after two doses, nor evidence that a low nadir CD4+ T-cell count compromised responses. Post-third-dose humoral responses substantially exceeded post-second-dose levels, though anti-Omicron responses were consistently weaker than against wild-type. Nevertheless, post-third-dose responses in PLWH were comparable to or higher than controls. An mRNA-1273 third dose was the strongest consistent correlate of higher post-third-dose responses. Conclusion: PLWH receiving suppressive antiretroviral therapy mount strong antibody responses after two- and three-dose COVID-19 vaccination. Results underscore the immune benefits of third doses in light of Omicron.
Collapse
Affiliation(s)
- Hope R Lapointe
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Francis Mwimanzi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Peter K Cheung
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Yurou Sang
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Fatima Yaseen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | | | - Rebecca Kalikawe
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Sarah Speckmaier
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | | | - Sneha Datwani
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Maggie C Duncan
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Olga Agafitei
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Siobhan Ennis
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Landon Young
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
| | - Hesham Ali
- John Ruedy Clinic, St, Paul's Hospital, Vancouver, Canada
| | - Bruce Ganase
- AIDS Research Program, St. Paul's Hospital, Vancouver, Canada
| | - F Harrison Omondi
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Winnie Dong
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Junine Toy
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Paul Sereda
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Laura Burns
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
| | - Cecilia T Costiniuk
- Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Curtis Cooper
- Department of Medicine, University of Ottawa, Ottawa, Canada.,Ottawa Hospital Research Institute, Ottawa, Canada
| | - Aslam H Anis
- School of Population and Public Health, University of British Columbia, Vancouver, Canada.,CIHR Canadian HIV Trials Network, University of British Columbia, Vancouver, Canada.,Centre for Health Evaluation and Outcome Sciences, Vancouver, Canada
| | - Victor Leung
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Daniel Holmes
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Mari L DeMarco
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Janet Simons
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Natalie Prystajecky
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,British Columbia Centre for Disease Control Public Health Laboratory, Vancouver, Canada
| | - Christopher F Lowe
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Ralph Pantophlet
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Marc G Romney
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Rolando Barrios
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Silvia Guillemi
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Department of Family Practice, Faculty of Medicine, University of British Columbia, Canada
| | - Chanson J Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Julio S G Montaner
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Mark Hull
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Department of Family Practice, Faculty of Medicine, University of British Columbia, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Mark A Brockman
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Zabrina L Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
7
|
Brumme ZL, Mwimanzi F, Lapointe HR, Cheung PK, Sang Y, Duncan MC, Yaseen F, Agafitei O, Ennis S, Ng K, Basra S, Lim LY, Kalikawe R, Speckmaier S, Moran-Garcia N, Young L, Ali H, Ganase B, Umviligihozo G, Omondi FH, Atkinson K, Sudderuddin H, Toy J, Sereda P, Burns L, Costiniuk CT, Cooper C, Anis AH, Leung V, Holmes D, DeMarco ML, Simons J, Hedgcock M, Romney MG, Barrios R, Guillemi S, Brumme CJ, Pantophlet R, Montaner JSG, Niikura M, Harris M, Hull M, Brockman MA. Humoral immune responses to COVID-19 vaccination in people living with HIV receiving suppressive antiretroviral therapy. NPJ Vaccines 2022; 7:28. [PMID: 35228535 PMCID: PMC8885829 DOI: 10.1038/s41541-022-00452-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/19/2022] [Indexed: 12/30/2022] Open
Abstract
Humoral responses to COVID-19 vaccines in people living with HIV (PLWH) remain incompletely characterized. We measured circulating antibodies against the SARS-CoV-2 spike protein receptor-binding domain (RBD), ACE2 displacement and viral neutralization activities one month following the first and second COVID-19 vaccine doses, and again 3 months following the second dose, in 100 adult PLWH and 152 controls. All PLWH were receiving suppressive antiretroviral therapy, with median CD4+ T-cell counts of 710 (IQR 525-935) cells/mm3, though nadir CD4+ T-cell counts ranged as low as <10 cells/mm3. After adjustment for sociodemographic, health and vaccine-related variables, HIV infection was associated with lower anti-RBD antibody concentrations and ACE2 displacement activity after one vaccine dose. Following two doses however, HIV was not significantly associated with the magnitude of any humoral response after multivariable adjustment. Rather, older age, a higher burden of chronic health conditions, and dual ChAdOx1 vaccination were associated with lower responses after two vaccine doses. No significant correlation was observed between recent or nadir CD4+ T-cell counts and responses to two vaccine doses in PLWH. These results indicate that PLWH with well-controlled viral loads and CD4+ T-cell counts in a healthy range generally mount strong initial humoral responses to dual COVID-19 vaccination. Factors including age, co-morbidities, vaccine brand, response durability and the rise of new SARS-CoV-2 variants will influence when PLWH will benefit from additional doses. Further studies of PLWH who are not receiving antiretroviral treatment or who have low CD4+ T-cell counts are needed, as are longer-term assessments of response durability.
Collapse
Affiliation(s)
- Zabrina L Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada.
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.
| | - Francis Mwimanzi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Hope R Lapointe
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Peter K Cheung
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Yurou Sang
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Maggie C Duncan
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Fatima Yaseen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Olga Agafitei
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Siobhan Ennis
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Kurtis Ng
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Simran Basra
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, Canada
| | - Li Yi Lim
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Rebecca Kalikawe
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Sarah Speckmaier
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | | | - Landon Young
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
| | - Hesham Ali
- John Ruedy Clinic, St, Paul's Hospital, Vancouver, Canada
| | - Bruce Ganase
- AIDS Research Program, St. Paul's Hospital, Vancouver, Canada
| | | | - F Harrison Omondi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Kieran Atkinson
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Hanwei Sudderuddin
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Junine Toy
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Paul Sereda
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Laura Burns
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
| | - Cecilia T Costiniuk
- Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Curtis Cooper
- Department of Medicine, University of Ottawa, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canada
| | - Aslam H Anis
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
- CIHR Canadian HIV Trials Network, University of British Columbia, Vancouver, Canada
- Centre for Health Evaluation and Outcome Sciences, Vancouver, Canada
| | - Victor Leung
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Daniel Holmes
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Mari L DeMarco
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Janet Simons
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Marc G Romney
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Rolando Barrios
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Silvia Guillemi
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Family Practice, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Chanson J Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Ralph Pantophlet
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Julio S G Montaner
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Family Practice, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Mark Hull
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Mark A Brockman
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
8
|
Brumme ZL, Mwimanzi F, Lapointe HR, Cheung P, Sang Y, Duncan MC, Yaseen F, Agafitei O, Ennis S, Ng K, Basra S, Lim LY, Kalikawe R, Speckmaier S, Moran-Garcia N, Young L, Ali H, Ganase B, Umviligihozo G, Omondi FH, Atkinson K, Sudderuddin H, Toy J, Sereda P, Burns L, Costiniuk CT, Cooper C, Anis AH, Leung V, Holmes D, DeMarco ML, Simons J, Hedgcock M, Romney MG, Barrios R, Guillemi S, Brumme CJ, Pantophlet R, Montaner JS, Niikura M, Harris M, Hull M, Brockman MA. Humoral immune responses to COVID-19 vaccination in people living with HIV receiving suppressive antiretroviral therapy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.10.03.21264320. [PMID: 34671779 PMCID: PMC8528088 DOI: 10.1101/2021.10.03.21264320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Humoral responses to COVID-19 vaccines in people living with HIV (PLWH) remain incompletely understood. We measured circulating antibodies against the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein, ACE2 displacement and live viral neutralization activities one month following the first and second COVID-19 vaccine doses in 100 adult PLWH and 152 controls. All PLWH were receiving suppressive antiretroviral therapy, with median CD4+ T-cell counts of 710 (IQR 525-935) cells/mm 3 . Nadir CD4+ T-cell counts ranged as low as <10 (median 280; IQR 120-490) cells/mm 3 . After adjustment for sociodemographic, health and vaccine-related variables, HIV infection was significantly associated with 0.2 log 10 lower anti-RBD antibody concentrations (p=0.03) and ∼11% lower ACE2 displacement activity (p=0.02), but not lower viral neutralization (p=0.1) after one vaccine dose. Following two doses however, HIV was no longer significantly associated with the magnitude of any response measured. Rather, older age, a higher burden of chronic health conditions, and having received two ChAdOx1 doses (versus a heterologous or dual mRNA vaccine regimen) were independently associated with lower responses. After two vaccine doses, no significant correlation was observed between the most recent or nadir CD4+ T-cell counts and vaccine responses in PLWH. These results suggest that PLWH with well-controlled viral loads on antiretroviral therapy and CD4+ T-cell counts in a healthy range will generally not require a third COVID-19 vaccine dose as part of their initial immunization series, though other factors such as older age, co-morbidities, vaccine regimen type, and durability of vaccine responses will influence when this group may benefit from additional doses. Further studies of PLWH who are not receiving antiretroviral treatment and/or who have low CD4+ T-cell counts are needed.
Collapse
Affiliation(s)
- Zabrina L. Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Francis Mwimanzi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Hope R. Lapointe
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Peter Cheung
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Yurou Sang
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Maggie C. Duncan
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Fatima Yaseen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Olga Agafitei
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Siobhan Ennis
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Kurtis Ng
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Simran Basra
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, Canada
| | - Li Yi Lim
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Rebecca Kalikawe
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Sarah Speckmaier
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | | | - Landon Young
- Division of Medical Microbiology and Virology, St. Paul’s Hospital, Vancouver, Canada
| | - Hesham Ali
- John Ruedy Clinic, St, Paul’s Hospital, Vancouver, Canada
| | - Bruce Ganase
- AIDS Research Program, St. Paul’s Hospital, Vancouver, Canada
| | | | - F. Harrison Omondi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Kieran Atkinson
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Hanwei Sudderuddin
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Junine Toy
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Paul Sereda
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Laura Burns
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
| | - Cecilia T. Costiniuk
- Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Curtis Cooper
- Department of Medicine, University of Ottawa, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, Canadas
| | - Aslam H. Anis
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
- CIHR Canadian HIV Trials Network, University of British Columbia, Vancouver, Canada
- Centre for Health Evaluation and Outcome Sciences, Vancouver, Canada
| | - Victor Leung
- Division of Medical Microbiology and Virology, St. Paul’s Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Daniel Holmes
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Mari L. DeMarco
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Janet Simons
- Department of Pathology and Laboratory Medicine, Providence Health Care, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Marc G. Romney
- Division of Medical Microbiology and Virology, St. Paul’s Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Rolando Barrios
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Silvia Guillemi
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Family Practice, Faculty of Medicine, University of British Columbia, Canada
| | - Chanson J. Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Ralph Pantophlet
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Julio S.G. Montaner
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Family Practice, Faculty of Medicine, University of British Columbia, Canada
| | - Mark Hull
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Mark A. Brockman
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
9
|
Bahemana E, Esber A, Dear N, Ganesan K, Parikh A, Reed D, Maganga L, Khamadi S, Mizinduko M, Lwilla A, Mkondoo D, Mwaisanga G, Somi N, Owouth J, Maswai J, Kiweewa F, Iroezindu M, Ake JA, Crowell TA, Valcour VG, Polyak CS. Impact of age on CD4 recovery and viral suppression over time among adults living with HIV who initiated antiretroviral therapy in the African Cohort Study. AIDS Res Ther 2020; 17:66. [PMID: 33183355 PMCID: PMC7664082 DOI: 10.1186/s12981-020-00323-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION With increased use of antiretroviral therapy (ART), HIV mortality rates are declining and people living with HIV (PLWH) are surviving longer. We characterized CD4 recovery and viral suppression among adults aged < 50 and ≥ 50 years living with HIV who initiated ART in the African Cohort Study (AFRICOS). METHODS Beginning in January 2013, PLWH at twelve clinics in Kenya, Uganda, Tanzania and Nigeria underwent medical history review, CD4 and viral load testing as part of the ongoing African Cohort Study (AFRICOS). ART-naïve PLWH who initiated ART within 30 days of enrollment and had at least one year of follow-up were included in these analyses. To compare ART response in participants < 50 years and ≥ 50 years old, changes in CD4 count and viral load suppression after ART initiation were examined at different time points using linear and binomial regression with generalized estimating equations. Variables for time since ART initiation and the interaction between age group and time on ART were included in the model to evaluate longitudinal changes in CD4 recovery and viral suppression by age. RESULTS Between January 2013 and September 2019, 2918 PLHV were enrolled in the cohort. Of these, 443 were ART naïve and initiated on ART within 30 days of enrollment, with 90% (n = 399) aged < 50 years old at ART initiation. At ART initiation, participants aged 50 and older had a higher median CD4 count compared to participants younger than 50 years of age although it did not reach statistical significance (306 cells/mm3, IQR:130-547 vs. 277cells/mm3, IQR: 132-437). In adjusted models examining CD4 recovery and viral suppression there were no significant differences by age group over time. By the end of follow-up viral suppression was high among both groups of adults (96% of adults ≥ 50 years old and 92% of adults < 50 years old). CONCLUSION This study found no difference in long-term CD4 recovery or viral suppression by age at ART initiation. We found that particularly among younger adults participants had lower median CD4 counts at ART initiation, suggesting the importance of identifying and putting this population on treatment earlier in the disease course.
Collapse
Affiliation(s)
- Emmanuel Bahemana
- HJF Medical Research International, Inc., Mbeya, Tanzania.
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - Allahna Esber
- 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.
| | - Nicole Dear
- 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
| | - Kavitha Ganesan
- 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
| | - Ajay Parikh
- 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
| | - Domonique Reed
- 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
| | - Lucas Maganga
- National Institute for Medical Research-Mbeya Medical Research Center, Mbeya, Tanzania
| | - Samoel Khamadi
- HJF Medical Research International, Inc., Mbeya, Tanzania
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Mucho Mizinduko
- Muhimbili University of Health and Allied Science-Dar-Es-Salaam, Dar-Es-Salaam, Tanzania
| | - Anange Lwilla
- HJF Medical Research International, Inc., Mbeya, Tanzania
| | - Dorothy Mkondoo
- HJF Medical Research International, Inc., Mbeya, Tanzania
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Gwamaka Mwaisanga
- HJF Medical Research International, Inc., Mbeya, Tanzania
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Nancy Somi
- HJF Medical Research International, Inc., Mbeya, Tanzania
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - John Owouth
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- HJF Medical Research International, Kisumu, Kenya
| | - Jonah Maswai
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- HJF Medical Research International, Kericho, Kenya
| | | | - Michael Iroezindu
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- HJF Medical Research International, Abuja, Nigeria
| | - Julie A Ake
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - 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
| | - Victor G Valcour
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Christina S Polyak
- 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
| |
Collapse
|
10
|
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: 11] [Impact Index Per Article: 2.2] [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
|
11
|
Acute HIV Infection and CD4/CD8 Ratio Normalization After Antiretroviral Therapy Initiation. J Acquir Immune Defic Syndr 2019; 79:510-518. [PMID: 30142143 DOI: 10.1097/qai.0000000000001843] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We estimated the effect of initiating virologically suppressive antiretroviral therapy (ART) during acute HIV infection versus chronic HIV infection (AHI vs. CHI) on CD4/CD8 ratio normalization. SETTING A prospective clinical cohort study. METHODS We included patients initiating ART with AHI and CHI between 2000 and 2015 and compared time from ART initiation to the first normal CD4/CD8 ratio (defined as CD4/CD8 ≥1) using Kaplan-Meier curves and multivariable Cox proportional hazards models. Patient time was censored at virologic failure, lost to follow-up, or death. We also characterized CD4, CD8, and CD4/CD8 trajectories over the first 3 years of ART. RESULTS The 1198 patients were 27% female and 60% African American, with a median age of 37 years (interquartile range 28-47) at ART initiation. The 83 AHI patients were more likely male, younger, and of white race, than CHI patients. After 2 years of suppressive ART, 70% of AHI patients achieved a normal CD4/CD8 ratio, compared to 6%-38% of CHI patients, with greater likelihood of normalization at higher baseline CD4 counts. Time to normalization was shortest among AHI patients, followed by CHI patients with higher baseline CD4. The adjusted hazard ratio for time to normalization for AHI patients compared to CHI patients with baseline CD4 >350 was 4.33 (95% CI: 3.16 to 5.93). Higher baseline CD4/CD8 ratio was also associated with time to normalization (adjusted hazard ratio 1.54; 1.46, 1.63, per 0.1 increase in ratio). CONCLUSIONS Initiating ART during AHI at higher baseline CD4 cell counts and CD4/CD8 ratios was associated with shorter time to CD4/CD8 ratio normalization.
Collapse
|
12
|
The benefit of immediate compared with deferred antiretroviral therapy on CD4+ cell count recovery in early HIV infection. AIDS 2019; 33:1335-1344. [PMID: 31157663 DOI: 10.1097/qad.0000000000002219] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess the impact of immediate vs. deferred antiretroviral therapy (ART) on CD4 recovery among individuals early in HIV infection. DESIGN Using serologic markers of early infection together with self-reported dates of infection and HIV diagnosis, ART-naive participants who were randomized to immediate vs. deferred ART in the Strategic Timing of Antiretroviral Treatment trial were classified into subgroups of duration of HIV infection at baseline. CD4 cell count recovery over follow-up according to duration of HIV infection was investigated. METHODS Three subgroups were defined: first, infected 6 months or less (n = 373); second, infected 6-24 months (n = 2634); and third, infected 24 months or longer (n = 1605). Follow-up CD4, CD8, and CD4 : CD8 ratio for the immediate and deferred ART groups were compared by subgroup using linear models. For the deferred ART group, decline to CD4 less than 350 cells/μl or AIDS according to infection duration was compared using time-to-event methods. RESULTS Follow-up CD4 cell count differences (immediate minus deferred) were greater for those recently infected (+231 cells/μl) compared with the two other subgroups (202 and 171 cells/μl; P < 0.001). CD4 : CD8 ratio treatment differences varied significantly (P < 0.001) according to duration of infection. In the deferred ART group, decline to CD4 less than 350 cells/μl or AIDS was greater among those recently infected (16.1 vs. 13.2 and 10.5 per 100 person years for those infected 6-24 and ≥24 months; P = 0.002). CONCLUSION In this randomized comparison of immediate vs. deferred ART, the CD4 cell count difference was greatest for those recently infected with HIV, emphasizing the importance of immediate ART initiation.
Collapse
|
13
|
SAHA SHUBHANKAR, ROY PRITIKUMAR, SMITH? ROBERT. MODELING MONOCYTE-DERIVED DENDRITIC CELLS AS A THERAPEUTIC VACCINE AGAINST HIV. J BIOL SYST 2019. [DOI: 10.1142/s0218339018500262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Successful immunologic control of HIV infection can be achieved in long-term non-progressors or HIV-1 controllers. Dendritic cells (DCs) are required for specific antigen presentation to naïve T lymphocytes and for antiviral, type I interferon secretion. To understand this mechanism, we develop a mathematical model that describes the role of direct presentation (replicating virus-infected DCs or other [Formula: see text] T cells directly) and cross presentation (DCs obtain antigen processed in other infected cells such as [Formula: see text] T lymphocytes) during HIV-1 infection. We find equilibria and determine stability in the case of no vaccination, and then, when vaccination is taken, we determine analytical thresholds for the strength and frequency of the vaccine to ensure the disease-free equilibrium remains stable. Our theoretical results suggest that the restoration of DC numbers may be predictive of immune restoration and may be a goal for immunotherapy to enhance viral control in a larger proportion of patients.
Collapse
Affiliation(s)
- SHUBHANKAR SAHA
- Centre for Mathematical Biology and Ecology, Department of Mathematics, Jadavpur University, Kolkata 700032, India
| | - PRITI KUMAR ROY
- Centre for Mathematical Biology and Ecology, Department of Mathematics, Jadavpur University, Kolkata 700032, India
| | - ROBERT SMITH?
- Department of Mathematics and Faculty of Medicine, The University of Ottawa, 585 King Edward Avenue, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
14
|
A majority of HIV persistence during antiretroviral therapy is due to infected cell proliferation. Nat Commun 2018; 9:4811. [PMID: 30446650 PMCID: PMC6240116 DOI: 10.1038/s41467-018-06843-5] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022] Open
Abstract
Antiretroviral therapy (ART) suppresses viral replication in people living with HIV. Yet, infected cells persist for decades on ART and viremia returns if ART is stopped. Persistence has been attributed to viral replication in an ART sanctuary and long-lived and/or proliferating latently infected cells. Using ecological methods and existing data, we infer that >99% of infected cells are members of clonal populations after one year of ART. We reconcile our results with observations from the first months of ART, demonstrating mathematically how a fossil record of historic HIV replication permits observed viral evolution even while most new infected cells arise from proliferation. Together, our results imply cellular proliferation generates a majority of infected cells during ART. Therefore, reducing proliferation could decrease the size of the HIV reservoir and help achieve a functional cure. HIV infected cells persist for decades in patients under ART, but the mechanisms responsible remain unclear. Here, Reeves et al. use modeling approaches adapted from ecology to show that cellular proliferation, rather than viral replication, generates a majority of infected cells during ART.
Collapse
|
15
|
CD32 expression is associated to T-cell activation and is not a marker of the HIV-1 reservoir. Nat Commun 2018; 9:2739. [PMID: 30013105 PMCID: PMC6048139 DOI: 10.1038/s41467-018-05157-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/12/2018] [Indexed: 11/26/2022] Open
Abstract
CD32 has been shown to be preferentially expressed in latently HIV-1-infected cells in an in vitro model of quiescent CD4 T cells. Here we show that stimulation of CD4+ T cells with IL-2, IL-7, PHA, and anti-CD3/CD28 antibodies induces T-cell proliferation, co-expression of CD32 and the activation of the markers HLA-DR and CD69. HIV-1 infection increases CD32 expression. 79.2% of the CD32+/CD4+ T cells from HIV+ individuals under antiretroviral treatment were HLA-DR+. Resting CD4+ T cells infected in vitro generally results in higher integration of provirus. We observe no difference in provirus integration or replication-competent inducible latent HIV-1 in CD32+ or CD32− CD4+ T cells from HIV+ individuals. Our results demonstrate that CD32 expression is a marker of CD4+ T cell activation in HIV+ individuals and raises questions regarding the immune resting status of CD32+ cells harboring HIV-1 proviruses. CD32 has been previously shown to be expressed preferentially by CD4 T cells latently harbouring HIV-1. Here the authors show that CD32 expression in CD4 T cells is associated with T cell activation, is up-regulated by HIV-1 infection and importantly does not appear to represent an enriched cellular niche for latent HIV-1.
Collapse
|
16
|
Loucif H, Gouard S, Dagenais-Lussier X, Murira A, Stäger S, Tremblay C, Van Grevenynghe J. Deciphering natural control of HIV-1: A valuable strategy to achieve antiretroviral therapy termination. Cytokine Growth Factor Rev 2018; 40:90-98. [PMID: 29778137 DOI: 10.1016/j.cytogfr.2018.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 02/06/2023]
Abstract
Antiretroviral therapy (ART) has dramatically reduced HIV-1-associated morbidity and mortality, and has transformed HIV-1 infection into a manageable chronic condition by suppressing viral replication. However, despite recent patient care improvements, ART still fails to cure HIV-1 infection due to the inability to counteract immune defects and metabolic disturbances that are associated with residual inflammation alongside viral persistence. Life-long drug administration also results in multiple side-effects in patients including lipodystrophy and insulin resistance. Thus, it is critical to find new ways to reduce the length of treatment and facilitate the termination of ART, for example by boosting protective immunity. The rare ability of some individuals to naturally control HIV-1 infection despite residual inflammation could be exploited to identify molecular mechanisms involved in host protection that may function as potential therapeutic targets. In this review, we highlight evidence illustrating the molecular and metabolic advantages of HIV-1 controllers over ART treated patients that contribute to the maintenance of effective antiviral immunity.
Collapse
Affiliation(s)
- Hamza Loucif
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, H7V 1B7, QC, Canada
| | - Steven Gouard
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, H7V 1B7, QC, Canada
| | - Xavier Dagenais-Lussier
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, H7V 1B7, QC, Canada
| | - Armstrong Murira
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, H7V 1B7, QC, Canada
| | - Simona Stäger
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, H7V 1B7, QC, Canada
| | - Cécile Tremblay
- Centre de Recherche de l'Université de Montréal, Montréal, QC, Canada
| | - Julien Van Grevenynghe
- Institut National de la Recherche Scientifique (INRS)-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, H7V 1B7, QC, Canada.
| |
Collapse
|
17
|
McGary CS, Deleage C, Harper J, Micci L, Ribeiro SP, Paganini S, Kuri-Cervantes L, Benne C, Ryan ES, Balderas R, Jean S, Easley K, Marconi V, Silvestri G, Estes JD, Sekaly RP, Paiardini M. CTLA-4 +PD-1 - Memory CD4 + T Cells Critically Contribute to Viral Persistence in Antiretroviral Therapy-Suppressed, SIV-Infected Rhesus Macaques. Immunity 2017; 47:776-788.e5. [PMID: 29045906 PMCID: PMC5679306 DOI: 10.1016/j.immuni.2017.09.018] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 06/13/2017] [Accepted: 09/27/2017] [Indexed: 01/09/2023]
Abstract
Antiretroviral therapy (ART) suppresses viral replication in HIV-infected individuals but does not eliminate the reservoir of latently infected cells. Recent work identified PD-1+ follicular helper T (Tfh) cells as an important cellular compartment for viral persistence. Here, using ART-treated, SIV-infected rhesus macaques, we show that CTLA-4+PD-1- memory CD4+ T cells, which share phenotypic markers with regulatory T cells, were enriched in SIV DNA in blood, lymph nodes (LN), spleen, and gut, and contained replication-competent and infectious virus. In contrast to PD-1+ Tfh cells, SIV-enriched CTLA-4+PD-1- CD4+ T cells were found outside the B cell follicle of the LN, predicted the size of the persistent viral reservoir during ART, and significantly increased their contribution to the SIV reservoir with prolonged ART-mediated viral suppression. We have shown that CTLA-4+PD-1- memory CD4+ T cells are a previously unrecognized component of the SIV and HIV reservoir that should be therapeutically targeted for a functional HIV-1 cure.
Collapse
Affiliation(s)
- Colleen S McGary
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Justin Harper
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Luca Micci
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Susan P Ribeiro
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sara Paganini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Clarisse Benne
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Emily S Ryan
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Sherrie Jean
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kirk Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA 30329, USA
| | - Vincent Marconi
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Rafick-Pierre Sekaly
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA.
| |
Collapse
|
18
|
Ding Y, Duan S, Wu Z, Ye R, Yang Y, Yao S, Wang J, Xiang L, Jiang Y, Lu L, Jia M, Detels R, He N. Timing of antiretroviral therapy initiation after diagnosis of recent human immunodeficiency virus infection and CD4(+) T-cell recovery. Clin Microbiol Infect 2015; 22:290.e5-8. [PMID: 26627338 DOI: 10.1016/j.cmi.2015.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/09/2015] [Accepted: 11/15/2015] [Indexed: 10/22/2022]
Abstract
We retrospectively examined the timing of antiretroviral therapy (ART) initiation and CD4(+) T-cell recovery over 36 months among recent human immunodeficiency virus (HIV) infections using BED (HIV-1 subtypes B, E and D) immunoglobulin G capture enzyme immunoassay (BED-CEIA). Regardless of baseline CD4(+) counts, individuals (n = 393) who initiated ART >2 months after diagnosis had significantly decreased probability and rate of achieving CD4(+) counts ≥900 cells/μL or ≥600 cells/μL than those individuals (n = 135) who started ART earlier (≤2 months). But the mean CD4(+) counts in two groups converged after 30 months of treatment. Early ART initiation leads to accelerated CD4(+) recovery, but does not offer a long-term advantage in CD4(+) counts.
Collapse
Affiliation(s)
- Y Ding
- Department of Epidemiology, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - S Duan
- Dehong Prefecture Centre for Disease Control and Prevention, Mangshi, Yunnan Province, China
| | - Z Wu
- National Centre for AIDS/STD Control and Prevention (NCAIDS), Chinese Centre for Disease Control and Prevention (China CDC), Beijing, China
| | - R Ye
- Dehong Prefecture Centre for Disease Control and Prevention, Mangshi, Yunnan Province, China
| | - Y Yang
- Dehong Prefecture Centre for Disease Control and Prevention, Mangshi, Yunnan Province, China
| | - S Yao
- Dehong Prefecture Centre for Disease Control and Prevention, Mangshi, Yunnan Province, China
| | - J Wang
- Dehong Prefecture Centre for Disease Control and Prevention, Mangshi, Yunnan Province, China
| | - L Xiang
- Dehong Prefecture Centre for Disease Control and Prevention, Mangshi, Yunnan Province, China
| | - Y Jiang
- National Centre for AIDS/STD Control and Prevention (NCAIDS), Chinese Centre for Disease Control and Prevention (China CDC), Beijing, China
| | - L Lu
- Yunnan Centre for Disease Control and Prevention, Kuming, Yunnan Province, China
| | - M Jia
- Yunnan Centre for Disease Control and Prevention, Kuming, Yunnan Province, China
| | - R Detels
- Department of Epidemiology, School of Public Health, University of California, Los Angeles, Los Angeles, USA
| | - N He
- Department of Epidemiology, School of Public Health, and the Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Sued O, Ambrosioni J, Nicolás D, Manzardo C, Agüero F, Claramonte X, Plana M, Tuset M, Pumarola T, Gallart T, Gatell JM, Miró JM. Structured Treatment Interruptions and Low Doses of IL-2 in Patients with Primary HIV Infection. Inflammatory, Virological and Immunological Outcomes. PLoS One 2015; 10:e0131651. [PMID: 26186440 PMCID: PMC4506046 DOI: 10.1371/journal.pone.0131651] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 06/03/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Interventions during primary HIV infection (PHI) can modify the clinical course during the chronic phase. The long-term effect of structured treatment interruptions (STI) followed by low doses of interleukin-2 (IL-2) in treated PHI patients is unknown. METHODS Twelve PHI patients with viral load (VL) <20 copies/mL, CD4 cells >500 cells/mm3, and CD4/CD8 ratio >1, on antiretroviral therapy (ART) initiated within the first 90 days of infection and continued for at least 12 months were included. They underwent four STI and were then allocated (week 0 of the study) to ART alone or ART plus low doses of IL-2. ART was stopped once VL <20 copies/mL ('final stop'). Primary endpoints were VL<3000 copies/mL and CD4 cells >500 cells/mm3 at 48 weeks; secondary endpoints were immune activation, inflammatory markers until 48 weeks and the time before resuming ART (CD4 <350 cells/mm3 or AIDS) after 'final stop', compared between groups. RESULTS Ten out of 12 patients were males, median age was 35 years and the main risk was men-who-have-sex-with-men. Only one out of 12 patients (in the STI group) maintained VL<3000 copies/mL and CD4 cells >500 cells/mm3 without ART at 48 weeks. All other virological and immunological parameters were comparable between groups at week 0, 'final stop' and week 48. However, the proportion of CD8-CD38+ cells, tumor necrosis factor and srIL-2 were higher in the IL-2 group at 'final stop' and week 24. All these differences vanished during follow-up. At 5 years after the final stop 3 out of 6 patients in the IL-2 group and 6 out of 6 patients in the STI group have resumed ART (P = 0.19). CONCLUSIONS STI and IL-2 failed to achieve virological control after ART interruption. STI were not deleterious in long-term follow-up, an important issue for eradication and functional cure trials. TRIAL REGISTRATION ClinicalTrials.gov NCT02300623.
Collapse
Affiliation(s)
- Omar Sued
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Juan Ambrosioni
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - David Nicolás
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Christian Manzardo
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Fernando Agüero
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Xavier Claramonte
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Montserrat Plana
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Montserrat Tuset
- Pharmacy Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Tomás Pumarola
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Teresa Gallart
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - José María Gatell
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
| | - José María Miró
- Infectious Diseases Service, Hospital Clínic–IDIBAPS, University of Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
20
|
Abstract
INTRODUCTION Highly active antiretroviral therapy has been the big paradigm for transforming HIV infection in a chronic disease. However, it requires lifelong administration as the HIV provirus integrated within infected cells cannot be eliminated and virus replication resumes following antiviral discontinuation. Cumulative toxicities, incomplete immune restoration, elevated cost, drug-drug interactions and selection of drug-resistant viruses are well-known limitations of prolonged HIV medication. AREAS COVERED The first proof-of-concept that HIV infection could be cured was the Berlin patient. By blocking infection of target cells, gene therapy may allow viral clearance from carriers or prevention of infection in newly exposed individuals. Advances in the field of gene-targeting strategies, T-cell-based approaches and human stem cells are revolutionizing the field. A series of ongoing and planned trials are testing gene therapy as HIV cure. The ultimate goal is the elimination of latent viral reservoirs in HIV-infected persons and the need for lifelong antiretroviral therapy. Following a search in PubMed, we have reviewed current gene therapy strategies investigated for HIV infection as well as the latest communications on HIV eradication presented at international conferences. EXPERT OPINION Multiple efforts are underway to reproduce the Berlin patient situation by engineering autologous T cells or hematopoietic stem cells resistant to HIV infection. There is no doubt that the major challenge is the elimination of latent viral reservoirs. With this goal in mind, we have entered a new era in the hope for HIV cure.
Collapse
Affiliation(s)
- Carmen de Mendoza
- Puerta de Hierro Research Institute and University Hospital, Department of Internal Medicine , Majadahonda, Madrid , Spain
| | | | | | | |
Collapse
|
21
|
Lodi S, Fisher M, Phillips A, De Luca A, Ghosn J, Malyuta R, Zangerle R, Moreno S, Vanhems P, Boufassa F, Guiguet M, Porter K. Symptomatic illness and low CD4 cell count at HIV seroconversion as markers of severe primary HIV infection. PLoS One 2013; 8:e78642. [PMID: 24244330 PMCID: PMC3828389 DOI: 10.1371/journal.pone.0078642] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 09/20/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The risk/benefit of initiating ART in primary HIV infection (PHI) is unclear. The benefits are more likely to outweigh the risks in patients with severe PHI. An accepted definition of severe PHI is, however, lacking. METHODS CASCADE patients with HIV test interval <6 months were classified as severe and non-severe PHI based on whether the following traits were recorded in the first 6 months following seroconversion: severe specific pre-defined symptoms, central nervous system-implicated illness, and ≥1, ≥2 CD4<350 (and <500) cells/mm(3). For each definition, we used Kaplan-Meier curves and Cox survival models to compare time to AIDS/death, censoring at the earlier of last clinic visit or 1/1/1997, when combination antiretroviral therapy (cART) became available. RESULTS Among 1108 included patients mostly males (85%) infected through sex between men (71%), 366 were diagnosed with AIDS/died. The risk of AIDS/death was significantly higher for individuals with severe symptoms, those with ≥1 CD4<350 cells/mm(3) or ≥2 CD4 <500 cells/mm(3) in the first 6 months [aHR (95% confidence interval) 2.1 (1.4,3.2), 2.0 (1.5,2.7), and 2.3, (1.5-3.5) respectively]. Median [interquantile range] survival for patients with ≥2, ≥1 and no CD4<350 cells/mm(3) within 6 months of seroconversion was 3.9 [2.7,6.5], 5.4 [4.5,8.4] and 8.1 [4.3,10.3] years, respectively. The diagnosis of CNS-implicated symptoms was rare and did not appear to be prognostic. CONCLUSION One CD4 count <350 or two <500 cells/mm(3) within 6 months of seroconversion and/or severe illness in PHI may be useful early indicators of individuals at high risk of disease progression.
Collapse
Affiliation(s)
- Sara Lodi
- Instituto de Salud Carlos III, Madrid, Spain
| | - Martin Fisher
- Brighton and Sussex University Hospitals National Health Service Trust, Brighton, United Kingdom
| | | | - Andrea De Luca
- University Division of Infectious Diseases, University Hospital of Siena, Siena, Italy
| | - Jade Ghosn
- Université Paris Descartes, EA 3620, Paris, France
| | - Ruslan Malyuta
- Perinatal Prevention of AIDS Initiative, Odessa, The Ukraine
| | | | | | - Philippe Vanhems
- Edouard Herriot Hospital, Lyon, and Universite' de Lyon 1, Lyon, France
| | - Faroudy Boufassa
- Inserm, CESP Centre for Research in Epidemiology and Population Health, U1018, Epidemiology of HIV and STI Team, Le Kremlin-Bicetre, France
| | | | - Kholoud Porter
- MRC Clinical Trials Unit at University College London, London, United Kingdom
| | | |
Collapse
|
22
|
The γδ T-cell receptor repertoire is reconstituted in HIV patients after prolonged antiretroviral therapy. AIDS 2013; 27:1557-62. [PMID: 23525030 DOI: 10.1097/qad.0b013e3283611888] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Determine whether reconstitution of Vγ2Vδ2 T cells in patients with HIV is due to new cell synthesis with recovery of the T-cell receptor repertoire or proliferative expansion of residual cells from the time of treatment initiation. DESIGN Perform a cross-sectional analysis of the T-cell receptor complexity of Vγ2 chain in patients treated for HIV, natural virus suppressors who control viremia to undetectable levels, patients with chronic low-level viremia in the absence of therapy, and uninfected controls. Apply quantitative methods for repertoire analysis to assess the degree of Vδ2 repertoire loss or reconstitution. METHODS T-cell receptor Vγ2 chain DNA clones (up to 300 per patient sample) were sequenced and aligned to enumerate the antigen-reactive subset with Vγ2-Jγ1.2 rearrangements. Predominant shared (public) sequences in each patient were compared to a reference library of public sequences from uninfected controls to assess the extent of similarity. Repertoire comparisons were quantified through bioinformatics testing. RESULTS Patients with prolonged virus suppression due to antiretroviral therapy reconstituted the Vγ2 T-cell repertoire to near-normal levels. Natural virus suppressors were similar to the treatment group. Severe defects in the Vγ2 T-cell receptor repertoire were observed in patients with chronic viremia despite the absence of overt disease. CONCLUSION Prolonged HIV suppression with antiretroviral therapy leads to reconstitution of the Vγ2Vδ2 T-cell subset deleted in HIV disease. Direct evidence for repair of the T-cell receptor repertoire supports a view that treatment-associated immune reconstitution is due to new cell synthesis and not to expansion of residual cell populations.
Collapse
|
23
|
Yao Y, Luo Y, He Y, Zheng Y, Zhang Q, Zhou H, Zeng S, Chen Z, He B, He M. The effect of a year of highly active antiretroviral therapy on immune reconstruction and cytokines in HIV/AIDS patients. AIDS Res Hum Retroviruses 2013; 29:691-7. [PMID: 23151174 DOI: 10.1089/aid.2012.0275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
To investigate the effect of a year of highly active antiretroviral therapy (HAART) on immune reconstruction and cytokine production in HIV/AIDS patients, 35 AIDS patients were recruited for HAART treatment and 35 healthy volunteers were assigned as controls. The dynamic changes in HIV load, blood T cell subset counts, as well as interleukin (IL)-12, interferon (IFN)-γ, and interferon-inducible protein-10 (IP-10) levels in AIDS patients were evaluated before HAART and at 6 and 12 months after therapy. Our results revealed that HIV virus load in HIV/AIDS patients was reduced below the detectable limit after patients received 6 months of HAART. CD3(+)CD4(+), CD4(+)CD45RA(+)62L(+), and CD4(+)CD45RO(+) T cells were found to be significantly decreased in HIV/AIDS patients compared to the healthy controls, but increased after HAART. CD3(+)CD8(+) and CD8(+)CD38(+) cells were found to be increased in HIV/AIDS patients but decreased after HAART. Plasma IL-12 and IFN-γ levels were lower but IP-10 level was higher in AIDS patients compared to controls. HAART significantly improved IL-12 and IFN- γ levels but reduced IP-10 level in AIDS patients (p<0.01). CD4(+)CD45RA(+)62L(+) and CD4(+)CD45RO(+) T cells were positively correlated with plasma IL-12/IFN-γ levels (p<0.05), but negatively correlated with plasma IP-10 level. However, CD3(+)CD8(+) cells were negatively correlated with plasma IL-12 and IFN-γ levels, but positively correlated with IP-10 level (p<0.05). HAART benefits HIV/AIDS patients by not only inhibiting virus replication but also by contributing to immune reconstruction, such as restoring subsets of T cells and adjusting cytokine production in HIV/AIDS patients.
Collapse
Affiliation(s)
- Yunhai Yao
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yan Luo
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yan He
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yuhuang Zheng
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Quan Zhang
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Huaying Zhou
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Si Zeng
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Zi Chen
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Bo He
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Mei He
- Department of Infectious Disease, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
24
|
Feuth T, Arends JE, Fransen JH, Nanlohy NM, van Erpecum KJ, Siersema PD, Hoepelman AIM, van Baarle D. Complementary role of HCV and HIV in T-cell activation and exhaustion in HIV/HCV coinfection. PLoS One 2013; 8:e59302. [PMID: 23555014 PMCID: PMC3598709 DOI: 10.1371/journal.pone.0059302] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 02/13/2013] [Indexed: 11/18/2022] Open
Abstract
Objectives To investigate whether T-cell activation and exhaustion is linked to HCV- and HIV disease parameters in HIV/HCV infected individuals, we studied T-cell characteristics in HIV/HCV coinfected patients and controls. Methods 14 HIV/HCV coinfected, 19 HCV monoinfected, 10 HIV monoinfected patients and 15 healthy controls were included in this cross-sectional study. Differences in expression of activation and exhaustion markers (HLA-DR, CD38, PD-1, Tim-3 and Fas) and phenotypic markers on CD4+ and CD8+ T-cells were analysed by flow cytometry and were related to HCV disease parameters (HCV-viremia, ALT and liver fibrosis). Results Frequencies of activated CD4+ and CD8+ T-cells were higher in HIV/HCV-coinfected compared to healthy controls and HCV or HIV mono-infected individuals. Coinfected patients also showed high expression of the exhaustion marker PD-1 and death receptor Fas. In contrast, the exhaustion marker Tim-3 was only elevated in HIV-monoinfected patients. T-cell activation and exhaustion were correlated with HCV-RNA, suggesting that viral antigen influences T-cell activation and exhaustion. Interestingly, increased percentages of effector CD8+ T-cells were found in patients with severe (F3–F4) liver fibrosis compared to those with no to minimal fibrosis (F0–F2). Conclusions HIV/HCV coinfected patients display a high level of T-cell activation and exhaustion in the peripheral blood. Our data suggest that T-cell activation and exhaustion are influenced by the level of HCV viremia. Furthermore, high percentages of cytotoxic/effector CD8+ T-cells are associated with liver fibrosis in both HCV monoinfected and HIV/HCV coinfected patients.
Collapse
Affiliation(s)
- Thijs Feuth
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Joop E. Arends
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Justin H. Fransen
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Department of Immunology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Nening M. Nanlohy
- Department of Immunology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Karel J. van Erpecum
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Peter D. Siersema
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Andy I. M. Hoepelman
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Debbie van Baarle
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Department of Immunology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
25
|
Hoxie JA, June CH. Novel cell and gene therapies for HIV. Cold Spring Harb Perspect Med 2012; 2:2/10/a007179. [PMID: 23028130 DOI: 10.1101/cshperspect.a007179] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Highly active antiretroviral therapy dramatically improves survival in HIV-infected patients. However, persistence of HIV in reservoirs has necessitated lifelong treatment that can be complicated by cumulative toxicities, incomplete immune restoration, and the emergence of drug-resistant escape mutants. Cell and gene therapies offer the promise of preventing progressive HIV infection by interfering with HIV replication in the absence of chronic antiviral therapy. Individuals homozygous for a deletion in the CCR5 gene (CCR5Δ32) are largely resistant to infection from R5-topic HIV-1 strains, which are most commonly transmitted. A recent report that an HIV-infected patient with relapsed acute myelogenous leukemia was effectively cured from HIV infection after transplantation of hematopoietic stem/progenitor cells (HSC) from a CCR5Δ32 homozygous donor has generated renewed interest in developing treatment strategies that target viral reservoirs and generate HIV resistance in a patient's own cells. Although the development of cell-based and gene transfer therapies has been slow, progress in a number of areas is evident. Advances in the fields of gene-targeting strategies, T-cell-based approaches, and HSCs have been encouraging, and a series of ongoing and planned trials to establish proof of concept for strategies that could lead to successful cell and gene therapies for HIV are under way. The eventual goal of these studies is to eliminate latent viral reservoirs and the need for lifelong antiretroviral therapy.
Collapse
Affiliation(s)
- James A Hoxie
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA.
| | | |
Collapse
|
26
|
Disease-modifying therapeutic concepts for HIV in the era of highly active antiretroviral therapy. J Acquir Immune Defic Syndr 2011; 58:297-303. [PMID: 21792065 DOI: 10.1097/qai.0b013e31822ccfcc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic HIV infection is associated with persistent immune activation and inflammation even among patients virologically suppressed on antiretroviral therapy for years. Chronic immune activation has been associated with poor outcomes--both AIDS-defining and non-AIDS-defining clinical events--and persistent CD4 T-cell depletion. The cause of chronic immune activation in well-controlled HIV infection is unknown. Proposed drivers include residual viral replication, microbial translocation, and coinfecting pathogens. Therapeutic interventions targeting immune activation are emerging, from approaches that interfere directly with activation and inflammatory pathways to those that prevent microbial translocation or decrease the availability of host target cells for the virus. In the context of the disappointing results of the interleukin-2 trials, the main challenges to developing these disease-modifying therapies include identifying an adequate target population and choosing surrogate endpoints that will provide positive proof-of-concept that the interventions will translate into long-term clinical benefit before embarking on large clinical endpoint trials.
Collapse
|
27
|
Thorborn GS, Pomeroy L, Ishohanni H, Peters BS, Vyakarnam A. Elevated effector cell sensitivity to Treg-cell suppression that is not associated with reduced Th17-cell expression distinguishes HIV+asymptomatic subjects from progressors. Eur J Immunol 2011; 42:138-46. [DOI: 10.1002/eji.201141426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 09/06/2011] [Accepted: 10/06/2011] [Indexed: 11/12/2022]
|
28
|
Early and prolonged antiretroviral therapy is associated with an HIV-1-specific T-cell profile comparable to that of long-term non-progressors. PLoS One 2011; 6:e18164. [PMID: 21483676 PMCID: PMC3071718 DOI: 10.1371/journal.pone.0018164] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 02/22/2011] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Intervention with antiretroviral treatment (ART) and control of viral replication at the time of HIV-1 seroconversion may curtail cumulative immunological damage. We have therefore hypothesized that ART maintenance over a very prolonged period in HIV-1 seroconverters could induce an immuno-virological status similar to that of HIV-1 long-term non-progressors (LTNPs). METHODOLOGY/PRINCIPAL FINDINGS We have investigated a cohort of 20 HIV-1 seroconverters on long-term ART (LTTS) and compared it to one of 15 LTNPs. Residual viral replication and reservoirs in peripheral blood, as measured by cell-associated HIV-1 RNA and DNA, respectively, were demonstrated to be similarly low in both cohorts. These two virologically matched cohorts were then comprehensively analysed by polychromatic flow cytometry for HIV-1-specific CD4(+) and CD8(+) T-cell functional profile in terms of cytokine production and cytotoxic capacity using IFN-γ, IL-2, TNF-α production and perforin expression, respectively. Comparable levels of highly polyfunctional HIV-1-specific CD4(+) and CD8(+) T-cells were found in LTTS and LTNPs, with low perforin expression on HIV-1-specific CD8(+) T-cells, consistent with a polyfunctional/non-cytotoxic profile in a context of low viral burden. CONCLUSIONS Our results indicate that prolonged ART initiated at the time of HIV-1 seroconversion is associated with immuno-virological features which resemble those of LTNPs, strengthening the recent emphasis on the positive impact of early treatment initiation and paving the way for further interventions to promote virological control after treatment interruption.
Collapse
|
29
|
Cobb A, Roberts LK, Palucka AK, Mead H, Montes M, Ranganathan R, Burkeholder S, Finholt JP, Blankenship D, King B, Sloan L, Harrod AC, Lévy Y, Banchereau J. Development of a HIV-1 lipopeptide antigen pulsed therapeutic dendritic cell vaccine. J Immunol Methods 2011; 365:27-37. [DOI: 10.1016/j.jim.2010.11.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/05/2010] [Indexed: 10/18/2022]
|
30
|
Antiretroviral therapy down-regulates innate antiviral response genes in patients with AIDS in sub-saharan Africa. J Acquir Immune Defic Syndr 2011; 55:428-38. [PMID: 20838227 DOI: 10.1097/qai.0b013e3181ef4963] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE HIV pathogenesis is characterized by destructive imbalances between virus-mediated immune damage, antiviral immune responses, and immune activation. We characterized the effects of successful antiretroviral therapy (ART) to identify the breadth and patterns of HIV-associated gene expression. METHODS In a prospective observational, longitudinal cohort study of 10 ART-naive Ugandans with AIDS (median 30 CD4/μL), we measured mRNA gene profiles in peripheral blood using Affymetrix U133_Plus2.0 microarrays at 0, 2, 4, 8, and 24 weeks after ART initiation. RESULTS We identified 160 mRNA transcripts that were consistently down-regulated and 48 that were up-regulated after ART at each point over 24 weeks based on linear regression modeling (adjusted P < 0.05), Of these 208 transcripts, approximately half represent heretofore unrecognized ART-responsive genes and one-third have no known function. The down-regulated genes with known function encoded mediators of innate antiviral responses, including antiviral restriction factors, pattern recognition receptors, and interferon response proteins, and mediators of immune activation, cellular proliferation, and apoptosis. CONCLUSIONS By using ART to block the viral stimulus, we identified transcripts involved in innate antiviral immunity, including antiviral restriction factors and pattern recognition receptors, that were not previously known to be induced by HIV infection.
Collapse
|
31
|
Zhou J, Sirisanthana T, Kiertiburanakul S, Chen YMA, Han N, Lim PL, Kumarasamy N, Choi JY, Merati TP, Yunihastuti E, Oka S, Kamarulzaman A, Phanuphak P, Lee CKC, Li PCK, Pujari S, Saphonn V, Law MG. Trends in CD4 counts in HIV-infected patients with HIV viral load monitoring while on combination antiretroviral treatment: results from The TREAT Asia HIV Observational Database. BMC Infect Dis 2010; 10:361. [PMID: 21182796 PMCID: PMC3022834 DOI: 10.1186/1471-2334-10-361] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Accepted: 12/23/2010] [Indexed: 11/24/2022] Open
Abstract
Background The aim of this study was to examine the relationship between trends in CD4 counts (slope) and HIV viral load (VL) after initiation of combination antiretroviral treatment (cART) in Asian patients in The TREAT Asia HIV Observational Database (TAHOD). Methods Treatment-naive HIV-infected patients who started cART with three or more and had three or more CD4 count and HIV VL tests were included. CD4 count slopes were expressed as changes of cells per microliter per year. Predictors of CD4 count slopes from 6 months after initiation were assessed by random-effects linear regression models. Results A total of 1676 patients (74% male) were included. The median time on cART was 4.2 years (IQR 2.5-5.8 years). In the final model, CD4 count slope was associated with age, concurrent HIV VL and CD4 count, disease stage, hepatitis B or C co-infection, and time since cART initiation. CD4 count continues to increase with HIV VL up to 20 000 copies/mL during 6-12 months after cART initiation. However, the HIV VL has to be controlled below 5 000, 4 000 and 500 copies/mL for the CD4 count slope to remain above 20 cells/microliter per year during 12-18, 18-24, and beyond 24 months after cART initiation. Conclusions After cART initiation, CD4 counts continued to increase even when the concurrent HIV VL was detectable. However, HIV VL needed to be controlled at a lower level to maintain a positive CD4 count slope when cART continues. The effect on long-term outcomes through the possible development of HIV drug resistance remains uncertain.
Collapse
Affiliation(s)
- Jialun Zhou
- National Centre in HIV Epidemiology and Clinical Research, The University of New South Wales, Sydney, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Silvera R, Stein D, Hutt R, Hagerty R, Daskalakis D, Valentine F, Marmor M. The Development and Implementation of an Outreach Program to Identify Acute and Recent HIV Infections in New York City. Open AIDS J 2010; 4:76-83. [PMID: 20386719 PMCID: PMC2852119 DOI: 10.2174/1874613601004010076] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Revised: 08/11/2009] [Accepted: 09/16/2009] [Indexed: 11/22/2022] Open
Abstract
Introduction: Since 2004, the authors have been operating First Call NYU, an outreach program to identify acute and recent HIV infections, also called primary HIV infections, among targeted at-risk communities in the New York City (NYC) metropolitan area. Materials and Methodology: First Call NYU employed mass media advertising campaigns, outreach to healthcare providers in NYC, and Internet-based efforts including search engine optimization (SEO) and Internet-based advertising to achieve these goals. Results: Between October 2004 and October 2008, 571 individuals were screened through this program, leading to 446 unique, in-person screening visits. 47 primary HIV infections, including 14 acute and 33 recent HIV infections, were identified. Discussion: Internet and traditional recruitment methods can be used to increase self-referrals for screening following possible exposure to HIV. Conclusion: Community education of at-risk groups, with the goal of increased self-diagnosis of possible acute HIV infection, may be a useful addition to traditional efforts to identify such individuals.
Collapse
Affiliation(s)
- Richard Silvera
- Department of Environmental Medicine, NYU School of Medicine, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Ganesan A, Chattopadhyay PK, Brodie TM, Qin J, Gu W, Mascola JR, Michael NL, Follmann DA, Roederer M. Immunologic and virologic events in early HIV infection predict subsequent rate of progression. J Infect Dis 2010; 201:272-84. [PMID: 20001854 PMCID: PMC2939466 DOI: 10.1086/649430] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Variability in human immunodeficiency virus (HIV) disease progression cannot be fully predicted by CD4(+) T cell counts or viral load (VL). Because central memory T (T(CM)) cells play a critical role in the pathogenesis of simian immunodeficiency virus disease, we hypothesized that quantifying these cells in early HIV infection could provide prognostic information. METHODS We measured expression of CD45RO, chemokine (C-C motif) receptor (CCR) 5, CCR7, CD27, and CD28 to enumerate naive and memory subsets in samples from recently infected individuals. We also quantified proliferation, CD127 expression, and cell-associated VL. Disease progression was compared across subgroups defined by these measurements, using Kaplan-Meier survival curves and multivariate Cox proportional hazards regression. RESULTS Four hundred sixty-six subjects contributed 101 events. The proportion or absolute count of T(CM) cells did not correlate with disease progression, defined as the time to AIDS or death. However, significant associations were observed for proliferation within CD4(+) or CD8(+) T cells, loss of naive or CD127(+) memory CD8(+) T cells, and CD4(+) T cell-associated VL. CONCLUSIONS Our results demonstrate that the extent of the immunopathogenesis established early in HIV infection predicts the course of future disease. Because antiretroviral drug treatment reverses such defects in part, our study provides mechanistic clues to why early use of antiretrovirals may prove beneficial.
Collapse
Affiliation(s)
- Anuradha Ganesan
- National Naval Medical Center, Infectious Disease Clinical Research Program, Uniformed Services University, Bethesda, MD
| | - Pratip K. Chattopadhyay
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | - Tess M. Brodie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | - Jing Qin
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | - Wenjuan Gu
- Biostatistics Research Branch, SAIC-Frederick, Inc, Frederick, MD
| | - John R. Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | - Nelson L. Michael
- United States Military HIV Research Program, Walter Reed Army Institute of Research, Rockville, MD
| | - Dean A. Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD
| |
Collapse
|
34
|
Cellerai C, Little SJ, Loes SKD. Treatment of acute HIV-1 infection: are we getting there? Curr Opin HIV AIDS 2009; 3:67-74. [PMID: 19372946 DOI: 10.1097/coh.0b013e3282f31d4b] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW Treatment of primary HIV-1 infection may alter the natural history of HIV-1 infection and delay the need for chronic antiretroviral therapy; it may also be a public health measure. We discuss the results of therapeutic trials and cohort studies, the occurrence of transmitted drug resistance, and recent findings in terms of immunopathogenesis and decay of viral reservoirs. RECENT FINDINGS Events at the time of primary HIV-1 infection are understood to set the scene for persistence of immunologic damage and chronic immune activation, with a rapid viral onslaught primarily on memory CD4 T cells at mucosal effector sites. The initiation of antiretroviral therapy at primary HIV-1 infection has been associated with a high degree of undetectable viremia in compliant patients and substantial decay of reservoirs in peripheral blood. The degree of immune reconstitution at the gut mucosal level, however, does not appear to be comparable to that in peripheral blood. SUMMARY Recent insights into the long-term consequences of the early burst of HIV-1 replication - together with transmitted drug resistance, onward transmission, and the possibility of decay of viral reservoirs - are important steps in helping to design future therapeutic strategies in primary HIV-1 infection in an era of intense drug and vaccine development.
Collapse
Affiliation(s)
- Cristina Cellerai
- Department of Immunology and Molecular Pathology, University College London, Royal Free Hospital Campus, London NW3 2QG, UK
| | | | | |
Collapse
|
35
|
Barqasho B, Nowak P, Tjernlund A, Kinloch S, Goh LE, Lampe F, Fisher M, Andersson J, Sönnerborg A. Kinetics of plasma cytokines and chemokines during primary HIV-1 infection and after analytical treatment interruption. HIV Med 2009; 10:94-102. [PMID: 19200172 DOI: 10.1111/j.1468-1293.2008.00657.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND There are strong theoretical arguments for initiating antiretroviral therapy (ART) during primary HIV-1 infection (PHI) to preserve HIV-1-specific T-cell responses and to decrease immune activation. METHODS We assessed the degree of immune activation during PHI and after analytical treatment interruption (ATI) in plasma samples from 22 subjects by measuring 13 cytokines/chemokines with the Luminex system. Subjects initiated quadruple ART at PHI (the QUEST cohort) and were classified as responders or nonresponders according to their HIV-1 viral load (VL) 6 months post-ATI. RESULTS During PHI, nonresponders had higher levels of HIV-1 RNA, interferon (IFN)-gamma, tumour necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-10 and eotaxin than responders (P</=0.05). A positive correlation was found between VL and IFN-alpha, TNF-alpha, IL-1beta, macrophage inflammatory protein (MIP)-1alpha and MIP-1beta, respectively. Post ATI, responders had higher levels of IFN-gamma, MIP-1beta and monocyte chemotactic protein (MCP)-1 than nonresponders, while nonresponders had higher levels of HIV-1 RNA, IL-15 and eotaxin. Cytokine/chemokine levels were higher during PHI than post-ATI. CONCLUSIONS High levels of immune activation during PHI are associated with a worse virological outcome post-ATI. In contrast, VL post-ATI is negatively correlated with IFN-gamma and chemokines. Therefore, the degree of immune activation during PHI is associated with both the VL at PHI and the viral set-point post-ART.
Collapse
Affiliation(s)
- B Barqasho
- Department of Laboratory Medicine, Division of Clinical Virology, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Long-term patterns in CD4 response are determined by an interaction between baseline CD4 cell count, viral load, and time: The Asia Pacific HIV Observational Database (APHOD). J Acquir Immune Defic Syndr 2009; 50:513-20. [PMID: 19408354 DOI: 10.1097/qai.0b013e31819906d3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Random effects models were used to explore how the shape of CD4 cell count responses after commencing combination antiretroviral therapy (cART) develop over time and, in particular, the role of baseline and follow-up covariates. METHODS Patients in Asia Pacific HIV Observational Database who first commenced cART after January 1, 1997, and who had a baseline CD4 cell count and viral load measure and at least 1 follow-up measure between 6 and 24 months, were included. CD4 cell counts were determined at every 6-month period after the commencement of cART for up to 6 years. RESULTS A total of 1638 patients fulfilled the inclusion criteria with a median follow-up time of 58 months. Lower post-cART mean CD4 cell counts were found to be associated with increasing age (P < 0.001), pre-cART hepatitis C coinfection (P = 0.038), prior AIDS (P = 0.019), baseline viral load < or equal to 100,000 copies per milliliter (P < 0.001), and the Asia Pacific region compared with Australia (P = 0.005). A highly significant 3-way interaction between the effects of time, baseline CD4 cell count, and post-cART viral burden (P < 0.0001) was demonstrated. Higher long-term mean CD4 cell counts were associated with lower baseline CD4 cell count and consistently undetectable viral loads. Among patients with consistently detectable viral load, CD4 cell counts seemed to converge for all baseline CD4 levels. CONCLUSIONS Our analysis suggest that the long-term shape of post-cART CD4 cell count changes depends only on a 3-way interaction between baseline CD4 cell count, viral load response, and time.
Collapse
|
37
|
Seng R, Goujard C, Desquilbet L, Sinet M, Rouzioux C, Deveau C, Boufassa F, Delfraissy JF, Meyer L, Venet A. Rapid CD4+ cell decrease after transient cART initiated during primary HIV infection (ANRS PRIMO and SEROCO cohorts). J Acquir Immune Defic Syndr 2008; 49:251-8. [PMID: 18845951 DOI: 10.1097/qai.0b013e318189a739] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To modelize the rate of CD4 cell count decline and its determinants after cessation of combination antiretroviral therapy (cART) started during primary HIV infection (PHI) and compare it with never-treated patients. METHODS Kinetics of CD4 counts were analyzed on the square root scale by using a mixed-effects model in 170 patients who received cART during PHI from the Primary Infection (PRIMO) cohort and 123 never-treated patients from the Seroconverters (SEROCO) cohort. RESULTS After cART interruption in the PRIMO cohort, the CD4 cell count fell rapidly during the first 5 months and more slowly thereafter. The timing of treatment initiation had no influence on the rate of CD4 cell decline. In contrast, a larger increase in CD4 cell counts during cART was associated with a steeper decline and a larger loss of CD4 cells after treatment interruption. The mean CD4 cell loss 3 years postinterruption was 383 cells per microliter. In the SEROCO cohort, the CD4 T-cell decline was less steep (3-year CD4 loss 239 cells/microL). As a result, the mean CD4 cell counts were similar (416 cells/microL) 3 years after cART interruption (PRIMO) or after infection (SEROCO). CONCLUSIONS These data question the benefit of a limited course of cART even when initiated within 3 months after PHI diagnosis.
Collapse
|
38
|
HIV testing in adolescents and young adults receiving STI testing in an urban primary care setting. Sex Transm Dis 2008; 35:686-8. [PMID: 18449070 DOI: 10.1097/olq.0b013e31816b1f9b] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Goujard C, Marcellin F, Hendel-Chavez H, Burgard M, Meiffrédy V, Venet A, Rouzioux C, Taoufik Y, El Habib R, Beumont-Mauviel M, Aboulker JP, Lévy Y, Delfraissy JF. Interruption of antiretroviral therapy initiated during primary HIV-1 infection: impact of a therapeutic vaccination strategy combined with interleukin (IL)-2 compared with IL-2 alone in the ANRS 095 Randomized Study. AIDS Res Hum Retroviruses 2007; 23:1105-13. [PMID: 17919105 DOI: 10.1089/aid.2007.0047] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
HIV-specific T cell responses play a critical role in the control of infection. We evaluated the impact of immune-based interventions in patients first treated during primary HIV-1 infection (PHI). Forty-three patients were randomized within three groups, to receive either interleukin-2 (IL-2 group), or boosts of ALVAC-HIV (vCP1433) and LIPO-6T followed by interleukin-2 (Vac-IL2 group), compared with no immune intervention (control group), and were monitored for T cell responses. Impact of strategies on viral replication was subsequently assessed during long-term treatment interruption. HIV-specific CD4(+) T cell responses did not change during the study period in immunized patients relative to controls, and vaccination had only a transient effect on interferon-gamma-producing CD8 responses. Viral rebound after treatment interruption was similar in immunized patients and controls. Forty percent of patients had HIV RNA values <10,000 copies/ml 12 weeks after interruption. The cumulative time off treatment represented almost half the total follow-up period. Immunological and virological status during PHI and HIV DNA load at interruption were predictive of the level of viral rebound after treatment interruption, whereas HIV RNA level during PHI and HIV DNA level at interruption were predictive of the time off treatment. Treatment interruption is safe in patients treated early after primary HIV infection. On the basis of this pilot study, HIV immunizations and interleukin-2 appear to have no supplementary benefit.
Collapse
Affiliation(s)
- Cécile Goujard
- Service de Médecine Interne, Bicêtre Hospital, AP-HP, Le Kremlin Bicêtre, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
HIV infection starts as an acute, systemic infection, followed by a chronic period of clinical latency, usually lasting 3 to 10 years, which precedes the eventual collapse of the immune system. It is increasingly recognized that events occurring during acute HIV infection may determine the natural course of the disease. The very dynamic events of acute HIV infection provide multiple opportunities for biologic interventions, such as anti-retroviral or immune-based therapies. Similarly, the implementation of public health measures during acute HIV infection could help control epidemics or outbreaks. Many of the dramatic possibilities for intervention in acute HIV infection remain unproved, not the least because of traditional difficulty of diagnosing patients during this early period. This article reviews the natural history, pathogenesis and clinical presentation of acute HIV infection, and suggests a diagnostic and therapeutic approach to guide clinicians dealing with patients with suspected or confirmed acute HIV infection.
Collapse
Affiliation(s)
- Nicola M Zetola
- Division of Infectious Diseases, University of California-San Francisco, San Francisco, CA 94103, USA
| | | |
Collapse
|
41
|
Vasan S, Poles MA, Horowitz A, Siladji EE, Markowitz M, Tsuji M. Function of NKT cells, potential anti-HIV effector cells, are improved by beginning HAART during acute HIV-1 infection. Int Immunol 2007; 19:943-51. [PMID: 17702988 DOI: 10.1093/intimm/dxm055] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
NKT cells are a subset of lymphocytes that share features of T cells and NK cells and bridge the innate and adaptive immune responses. They are able to be infected by HIV, but their function in HIV-infected individuals is not known. NKT cell percentage and function was measured in individuals with acute HIV infection before and 1 year into highly active anti-retroviral therapy (HAART). This study demonstrates that percentages of both CD161+ NKT cells and CD161+, CD4+ NKT cells decline within the first few months after HIV-1 infection, but initiating therapy during the acute infection period can prevent a further decline in these NKT cell subsets during the first year. NKT cell function is also impaired during early HIV infection, but significantly improved by effective treatment with HAART. Finally, preservation of NKT cell function may be important in HIV-infected individuals, as NKT cells display an anti-HIV-1 activity in vitro, mediated by IFN-gamma secretion.
Collapse
Affiliation(s)
- Sandhya Vasan
- Aaron Diamond AIDS Research Center, Rockefeller University, 455 First Avenue, 7th Floor, New York, NY 10016, USA
| | | | | | | | | | | |
Collapse
|
42
|
Gras L, Kesselring AM, Griffin JT, van Sighem AI, Fraser C, Ghani AC, Miedema F, Reiss P, Lange JMA, de Wolf F. CD4 cell counts of 800 cells/mm3 or greater after 7 years of highly active antiretroviral therapy are feasible in most patients starting with 350 cells/mm3 or greater. J Acquir Immune Defic Syndr 2007; 45:183-92. [PMID: 17414934 DOI: 10.1097/qai.0b013e31804d685b] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE CD4 cell count changes in therapy-naive patients were investigated during 7 years of highly active antiretroviral therapy (HAART) in an observational cohort. METHODS Three endpoints were studied: (1) time to >or=800 CD4 cells/mm in 5299 therapy-naive patients starting HAART, (2) CD4 cell count changes during 7 years of uninterrupted HAART in a subset of 544 patients, and (3) reaching a plateau in CD4 cell restoration after 5 years of HAART in 366 virologically suppressed patients. RESULTS Among patients with <50, 50 to 200, 200 to 350, 350 to 500, and >or=500 CD4 cells/mm at baseline, respectively, 20%, 26%, 46%, 73%, and 87% reached >or=800 CD4 cells/mm within 7 years of starting HAART. Periods with HIV RNA levels >500 copies/mL and age >or=50 years were associated with lesser increases in CD4 cell counts between 6 months and 7 years. Having reached >or=800 CD4 cells/mm at 5 years, age >or=50 years, and >or=1 HIV RNA measurement >1000 copies/mL between 5 and 7 years were associated with a plateau in CD4 cell restoration. CONCLUSIONS Restoration to CD4 cell counts >or=800 cells/mm is feasible within 7 years of HAART in most HIV-infected patients starting with >or=350 cells/mm and achieving sufficient suppression of viral replication. Particularly in patients >or=50 years of age, it may be beneficial to start earlier than current guidelines recommend.
Collapse
Affiliation(s)
- Luuk Gras
- HIV Monitoring Foundation, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hoen B, Cooper DA, Lampe FC, Perrin L, Clumeck N, Phillips AN, Goh LE, Lindback S, Sereni D, Gazzard B, Montaner J, Stellbrink HJ, Lazzarin A, Ponscarme D, Staszewski S, Mathiesen L, Smith D, Finlayson R, Weber R, Wegmann L, Janossy G, Kinloch-de Loes S. Predictors of virological outcome and safety in primary HIV type 1-infected patients initiating quadruple antiretroviral therapy: QUEST GW PROB3005. Clin Infect Dis 2007; 45:381-90. [PMID: 17599319 DOI: 10.1086/519428] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 04/03/2007] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Initiation of antiretroviral therapy during primary human immunodeficiency virus (HIV)-1 infection may confer long-term benefit. METHODS After initiation of zidovudine, lamivudine, abacavir, and amprenavir therapy in patients in the QUEST cohort, predictors of virological outcome, virological and immunological changes, and adverse events were evaluated over 48 weeks. RESULTS One hundred forty-eight patients started antiretroviral therapy during primary HIV-1 infection with < or =3 bands on Western Blot (median plasma HIV-1 RNA load, 5.4 log copies/mL; median CD4 cell count, 517 cells/mm(3)). By week 48, 36% of patients had stopped treatment or were lost to follow-up. Among the 115 patients receiving follow-up care at week 48 (102 of whom were receiving antiretroviral therapy), the median viral load decrease was -5.4 log copies/mL (interquartile range [IQR], -6.4 to -3.9 log copies/mL), and the median increase in CD4 cell count was 147 cells/mm(3) (IQR, -1 to 283 cells/mm(3)); 84.2% of patients had a viral load < or =50 copies/mL, and 44.7% of patients had a viral load < or =3 copies/mL. The median cell-associated RNA level decreased from 3.4 log copies/million PBMCs (IQR, 2.9-4.1 log copies/million PBMCs) to 0.8 log copies/million PBMCs (IQR, 0.5-1.4 log copies/million PBMCs), and the median cell-associated DNA level decreased from 2.8 log copies/million PBMCs (IQR, 2.4-3.0 log copies/million PBMCs) to 1.6 log copies/million PBMCs (IQR, 1.2-1.9 log copies/million PBMCs); 33.3% of patients had an undetectable RNA level, and 9.5% of patients had an undetectable cell-associated DNA level. The median CD8(+)/CD38(++) T cell count decreased from 459 cells/mm(3) (IQR, 208-974 cells/mm(3)) to 33 cells/mm(3) (IQR, 19-75 cells/mm(3)). Baseline CD8(+)/CD38(++) T cell count and cell-associated DNA level were independent inverse predictors for reaching a viral load < or =3 copies/mL. Eighty-three patients experienced a serious adverse event (median duration of an adverse event, 15 days).Conclusions. Initiation of antiretroviral therapy during primary HIV-1 infection was associated with very significant antiretroviral activity and a decrease in immune activation. Lower baseline CD8(+)/CD38(++) T cell count and cell-associated DNA level were predictive of achieving a viral load < or =3 copies/mL.
Collapse
Affiliation(s)
- Bruno Hoen
- Department of Infectious Diseases, University Medical Centre, Besancon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Mathematical modeling is becoming established in the immunologist's toolbox as a method to gain insight into the dynamics of the immune response and its components. No more so than in the case of the study of human immunodeficiency virus (HIV) infection, where earlier work on the viral dynamics brought significant advances in our understanding of HIV replication and evolution. Here, I review different areas of the study of the dynamics of CD4+ T cells in the setting of HIV, where modeling played important and diverse roles in helping us understand CD4+ T-cell homeostasis and the effect of HIV infection. As the experimental techniques become more accurate and quantitative, modeling should play a more important part in both experimental design and data analysis.
Collapse
Affiliation(s)
- Ruy M Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| |
Collapse
|
45
|
Abstract
The current literature is controversial in providing evidence to determine the optimal time to initiate therapy among patients with HIV. However, there is evidence that initiating early treatment might provide benefits by treating primary HIV infection, preserving normal immune function, suppressing HIV viral replication, deferring clinical progression, and reducing HIV transmission. The biggest challenges in initiating treatment early are issues related with long-term management, including toxicities, adherence, and drug resistance. However, the availability of superior new antiretroviral drugs and simplified regimens, the development of effective treatment strategy, and further improvement of adherence through directly observed treatment are addressing the issues and changing the balance towards earlier treatment.
Collapse
Affiliation(s)
- Cunlin Wang
- Medical College of Virginia Campus, Virginia Commonwealth University, 1000 East Clay Street, Richmond, VA 23298, USA.
| | | | | |
Collapse
|
46
|
Adalid-Peralta L, Grangeot-Keros L, Rudent A, Ngo-Giang-Huong N, Krzysiek R, Goujard C, Deveau C, Le Gall M, Meyer L, Emilie D, Rouzioux C. Impact of highly active antiretroviral therapy on the maturation of anti-HIV-1 antibodies during primary HIV-1 infection. HIV Med 2006; 7:514-9. [PMID: 17105510 DOI: 10.1111/j.1468-1293.2006.00406.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To study the impact of highly active antiretroviral therapy (HAART) on isotype switching and avidity maturation of HIV-1-specific immunoglobulin G (IgG) in patients with primary HIV-1 infection (PHI). METHODS We studied the emergence and the evolution of anti-HIV IgG antibodies by quantitative immunoblotting to analyse IgG subclasses and IgG avidity. Serum samples were obtained from 16 PHI patients from the French PRIMO Cohort Study at various points in the first year of infection: eight patients received no treatment (group I), and eight patients received efficient HAART (group II) during the study period. RESULTS Early initiation of HAART in PHI patients partially prevented an increase in anti-HIV-1 IgG levels. Within IgG subclasses, the amount of anti-HIV-1 IgG1 gradually increased with time in both groups, although levels remained lower in treated patients. The anti-p24 IgG2 level was always lower in group II. We observed a decrease in anti-p24 IgG3 over time in both groups. Treatment did not affect the maturation of HIV-1 IgG avidity, which increased in both groups until month 3 and then remained high until the end of the 12-month follow-up period. CONCLUSIONS HAART in PHI partially prevents the emergence of HIV-1 IgG antibodies, but does not affect the quality of these antibodies, as reflected in their isotype and avidity.
Collapse
Affiliation(s)
- L Adalid-Peralta
- INSERM U764, Institut Fédératif de Recherches 13, Clamart, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Bongiovanni M, Bini T, Casana M, Cicconi P, Tordato F, Monforte AD. Long-term immunologic outcome in HAART-experienced subjects receiving lopinavir/ritonavir. AIDS Res Hum Retroviruses 2006; 22:1096-105. [PMID: 17147495 DOI: 10.1089/aid.2006.22.1096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The long-term immunological efficacy of regimens including lopinavir/ritonavir (LPV/r) has not been assessed in HIV-infected HAART-experienced subjects. The present study included 452 consecutive HIV-infected outpatients starting LPV/r before May 2003 after failing (HIV-RNA > 1000 copies/ml) HAART. Four groups were considered according to CD4 cell counts at LPV/r initiation: group 1 (G1, n = 115) < 100 cells/mm(3); group 2 (G2, n = 113) 100-199 cells/mm(3); group 3 (G3, n = 115) 200-349 cells/mm(3); group 4 (G4, n = 109) >/= 350 cells/mm(3). The majority of patients were males (n = 320, 70.8%), the median age was 38 years, and 180 (39.6%) were on CDC stage C. The median time of previous HAART was 51.1 months (12-81.7) and a median of 7 antiretroviral regimens and of 3 protease inhibitors was changed before LPV/r. The mean CD4 cell count increase was 105, 113, 128, and 144 cells/mm(3) after 12 months (p < 0.01 for each group) and 128, 106, 90, and 100 cells/mm(3) at month 48 (p < 0.01 for each group) in G1, G2, G3, and G4, respectively. The mean increase was comparable among the four groups. The on treatment analysis showed a better immunologic response among G1 and G2 patients from month 36. Forty-seven patients (10.4%), mainly in G1 and G2, maintained LPV/r despite persistent HIV-RNA > 1000 copies/ml. A mean increase of 64 and 65 cells/mm(3) and of 88 and 56 cells/mm(3) at month 12 and 48 was observed in G1 and G2, respectively. The use of LPV/r-based regimens also provided a durable immunologic recovery in highly pretreated HIV-infected subjects.
Collapse
Affiliation(s)
- Marco Bongiovanni
- Clinic of Infectious Diseases, San Paolo Hospital, University of Milan, Milan, Italy.
| | | | | | | | | | | |
Collapse
|
48
|
Considerations in the design of randomized controlled trials evaluating the optimal time to initiate antiretroviral therapy in previously untreated HIV-1-infected patients. Curr Opin HIV AIDS 2006; 1:488-94. [PMID: 19372851 DOI: 10.1097/coh.0b013e328010f238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW This paper addresses design issues around 'when to start' trials in adults and children and in chronic and acute HIV infection. Issues for young children treated soon after birth and recently seroconverting adults differ, and are also discussed. RECENT FINDINGS No trials have been published addressing this question for combination antiretroviral therapy in chronic HIV infection, and the evidence base for guidelines has largely been derived from observational studies. The biases inherent in observational data and recent results from the adult SMART trial may have moved the level of equipoise from 'unstable certainty' (mitigating against randomization) to a renewed call for a very large clinical endpoint trial of when to start therapy in the era of highly active antiretroviral therapy. Older children could also be enrolled in such a trial, as the predictive value of CD4 cell count values in children over 5 years and young adults. In acute infection, ongoing trials in adults and children is similar considering strategies of early short-term combination antiretroviral therapy compared with deferred therapy. Although not strictly 'when to start trials', these approaches could reduce life-long exposure to antiretroviral therapy while maintaining clinical and immunological well being. SUMMARY Issues in the design of a future randomized trial of when to start antiretroviral therapy in chronic HIV infected adults and older children are discussed, while the role of early limited therapy in primary infection awaits results of ongoing trials.
Collapse
|
49
|
Scadden DT, Muse VV, Hasserjian RP. Case records of the Massachusetts General Hospital. Case 30-2006. A 41-year-old man with dyspnea, fever, and lymphadenopathy. N Engl J Med 2006; 355:1358-68. [PMID: 17005954 DOI: 10.1056/nejmcpc069021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- David T Scadden
- Department of Hematology-Oncology, Massachusetts General Hospital, USA
| | | | | |
Collapse
|
50
|
Almeida M, Cordero M, Almeida J, Orfao A. Persistent abnormalities in peripheral blood dendritic cells and monocytes from HIV-1-positive patients after 1 year of antiretroviral therapy. J Acquir Immune Defic Syndr 2006; 41:405-15. [PMID: 16652047 DOI: 10.1097/01.qai.0000209896.82255.d3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Antiretroviral therapy (ART) has led to marked decreases in morbidity and mortality rates among HIV-1-positive patients; however, immune recovery is not complete. Although dendritic cells (DCs) were shown to be involved in HIV-1 pathogenesis, few studies have investigated the effect of ART on DCs. We have analyzed the effect of ART on numerical distribution, expression of chemokine receptors, and ex vivo production of inflammatory cytokines by peripheral blood (PB) monocytes and DCs in a cohort of chronically infected HIV-1-positive patients. Patients were tested before therapy and at weeks +2, +4, +8, +12, and +52 after starting ART.Our results show an incomplete T-cell immune reconstitution in chronically infected patients who had undetectable plasma viremia while taking ART for 1 year. This was associated with persistent abnormalities at week +52 of ART, corresponding to increased numbers of CD16 DCs and monocytes, as well as altered expression of CXC chemokine receptors, in the form of increased CXCR1 expression on monocytes and decreased reactivity for CXCR2 and/or CXCR4 on myeloid and plasmacytoid DCs. In addition, an abnormally high spontaneous ex vivo secretion of inflammatory cytokines by CD16 DCs and monocytes was still detected after 1 year of ART. These abnormalities were especially pronounced in patients with less than 200 CD4 T cells/microL, which could be related to the persistence of undetected viral replication and sustained immune activation.
Collapse
Affiliation(s)
- Maria Almeida
- Servicio General de Citometría and Centro de Investigación del Cáncer, Universidad de Salamanca, Spain
| | | | | | | |
Collapse
|