51
|
Song W, He D, Brill I, Malhotra R, Mulenga J, Allen S, Hunter E, Tang J, Kaslow RA. Disparate associations of HLA class I markers with HIV-1 acquisition and control of viremia in an African population. PLoS One 2011; 6:e23469. [PMID: 21858133 PMCID: PMC3157381 DOI: 10.1371/journal.pone.0023469] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 07/18/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Acquisition of human immunodeficiency virus type 1 (HIV-1) infection is mediated by a combination of characteristics of the infectious and the susceptible member of a transmission pair, including human behavioral and genetic factors, as well as viral fitness and tropism. Here we report on the impact of established and potential new HLA class I determinants of heterosexual HIV-1 acquisition in the HIV-1-exposed seronegative (HESN) partners of serodiscordant Zambian couples. METHODOLOGY/PRINCIPAL FINDINGS We assessed the relationships of behavioral and clinically documented risk factors, index partner viral load, and host genetic markers to HIV-1 transmission among 568 cohabiting couples followed for at least nine months. We genotyped subjects for three classical HLA class I genes known to influence immune control of HIV-1 infection. From 1995 to December 2006, 240 HESNs seroconverted and 328 remained seronegative. In Cox proportional hazards models, HLA-A*68:02 and the B*42-C*17 haplotype in HESN partners were significantly and independently associated with faster HIV-1 acquisition (relative hazards = 1.57 and 1.55; p = 0.007 and 0.013, respectively) after controlling for other previously established contributing factors in the index partner (viral load and specific class I alleles), in the HESN partner (age, gender), or in the couple (behavioral and clinical risk score). Few if any previously implicated class I markers were associated here with the rate of acquiring infection. CONCLUSIONS/SIGNIFICANCE A few HLA class I markers showed modest effects on acquisition of HIV-1 subtype C infection in HESN partners of discordant Zambian couples. However, the striking disparity between those few markers and the more numerous, different markers found to determine HIV-1 disease course makes it highly unlikely that, whatever the influence of class I variation on the rate of infection, the mechanism mediating that phenomenon is identical to that involved in disease control.
Collapse
Affiliation(s)
- Wei Song
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Dongning He
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ilene Brill
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Rakhi Malhotra
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | | - Susan Allen
- Rwanda-Zambia HIV-1 Research Group, Lusaka, Zambia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Eric Hunter
- Vaccine Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Jianming Tang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Richard A. Kaslow
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
52
|
Ghunaim H, Kumar A, Torres J, Diaz-Mitoma F, Azizi A. An immunological comparison between lipidated and non-lipidated multivalent HIV-1 peptides representing Gp120 and Gag hypervariable regions. Vaccine 2011; 29:5950-8. [DOI: 10.1016/j.vaccine.2011.06.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 06/08/2011] [Accepted: 06/14/2011] [Indexed: 10/18/2022]
|
53
|
Girard MP, Osmanov S, Assossou OM, Kieny MP. Human immunodeficiency virus (HIV) immunopathogenesis and vaccine development: a review. Vaccine 2011; 29:6191-218. [PMID: 21718747 DOI: 10.1016/j.vaccine.2011.06.085] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/20/2011] [Accepted: 06/22/2011] [Indexed: 02/08/2023]
Abstract
The development of a safe, effective and globally affordable HIV vaccine offers the best hope for the future control of the HIV-1 pandemic. Since 1987, scores of candidate HIV-1 vaccines have been developed which elicited varying degrees of protective responses in nonhuman primate models, including DNA vaccines, subunit vaccines, live vectored recombinant vaccines and various prime-boost combinations. Four of these candidate vaccines have been tested for efficacy in human volunteers, but, to the exception of the recent RV144 Phase III trial in Thailand, which elicited a modest but statistically significant level of protection against infection, none has shown efficacy in preventing HIV-1 infection or in controlling virus replication and delaying progression of disease in humans. Protection against infection was observed in the RV144 trial, but intensive research is needed to try to understand the protective immune mechanisms at stake. Building-up on the results of the RV144 trial and deciphering what possibly are the immune correlates of protection are the top research priorities of the moment, which will certainly accelerate the development of an highly effective vaccine that could be used in conjunction with other HIV prevention and treatment strategies. This article reviews the state of the art of HIV vaccine development and discusses the formidable scientific challenges met in this endeavor, in the context of a better understanding of the immunopathogenesis of the disease.
Collapse
Affiliation(s)
- Marc P Girard
- University Paris 7, French National Academy of Medicine, 39 rue Seignemartin, FR 69008 Lyon, France.
| | | | | | | |
Collapse
|
54
|
Excler JL, Rida W, Priddy F, Gilmour J, McDermott AB, Kamali A, Anzala O, Mutua G, Sanders EJ, Koff W, Berkley S, Fast P. AIDS vaccines and preexposure prophylaxis: is synergy possible? AIDS Res Hum Retroviruses 2011; 27:669-80. [PMID: 21043994 PMCID: PMC3101085 DOI: 10.1089/aid.2010.0206] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
While the long-term goal is to develop highly effective AIDS vaccines, first generation vaccines may be only partially effective. Other HIV prevention modalities such as preexposure prophylaxis with antiretrovirals (PrEP) may have limited efficacy as well. The combined administration of vaccine and PrEP (VAXPREP), however, may have a synergistic effect leading to an overall benefit that is greater than the sum of the individual effects. We propose two test-of-concept trial designs for an AIDS vaccine plus oral or topical ARV. In one design, evidence that PrEP reduces the risk of HIV acquisition is assumed to justify offering it to all participants. A two-arm study comparing PrEP alone to VAXPREP is proposed in which 30 to 60 incident infections are observed to assess the additional benefit of vaccination on risk of infection and setpoint viral load. The demonstrated superiority of VAXPREP does not imply vaccine alone is efficacious. Similarly, the lack of superiority does not imply vaccine alone is ineffective, as antagonism could exist between vaccine and PrEP. In the other design, PrEP is assumed not to be in general use. A 2 × 2 factorial design is proposed in which high-risk individuals are randomized to one of four arms: placebo vaccine given with placebo PrEP, placebo vaccine given with PrEP, vaccine given with placebo PrEP, or VAXPREP. Between 60 and 210 infections are required to detect a benefit of vaccination with or without PrEP on risk of HIV acquisition or setpoint viral load, with fewer infections needed when synergy is present.
Collapse
Affiliation(s)
| | - Wasima Rida
- Biostatistics Consultant, Arlington, Virginia
| | - Frances Priddy
- International AIDS Vaccine Initiative, New York, New York
| | - Jill Gilmour
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | | | - Anatoli Kamali
- Medical Research Council, Uganda Virus Research Institute, Uganda Research Unit on AIDS, Entebbe, Uganda
| | - Omu Anzala
- Kenya AIDS Vaccine Initiative, University of Nairobi, Nairobi, Kenya
| | - Gaudensia Mutua
- Kenya AIDS Vaccine Initiative, University of Nairobi, Nairobi, Kenya
| | - Eduard J. Sanders
- Centre for Geographic Medicine Research—Coast, Kenya Medical Research Institute (KEMRI), Kilifi, Kenya
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Headington, United Kingdom
| | - Wayne Koff
- International AIDS Vaccine Initiative, New York, New York
| | - Seth Berkley
- International AIDS Vaccine Initiative, New York, New York
| | - Patricia Fast
- International AIDS Vaccine Initiative, New York, New York
| |
Collapse
|
55
|
Ritchie AJ, Campion SL, Kopycinski J, Moodie Z, Wang ZM, Pandya K, Moore S, Liu MKP, Brackenridge S, Kuldanek K, Legg K, Cohen MS, Delwart EL, Haynes BF, Fidler S, McMichael AJ, Goonetilleke N. Differences in HIV-specific T cell responses between HIV-exposed and -unexposed HIV-seronegative individuals. J Virol 2011; 85:3507-16. [PMID: 21270166 PMCID: PMC3067859 DOI: 10.1128/jvi.02444-10] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 01/17/2011] [Indexed: 11/20/2022] Open
Abstract
HIV-1-specific T lymphocyte responses in individuals exposed to HIV-1 but who remain persistently seronegative (HESNs) have been reported in some but not all previous studies. This study was designed to resolve unequivocally the question of whether HESNs make HIV-1-specific T cell responses. We performed a blind investigation to measure HIV-1-specific T cell responses in both HIV-1-serodiscordant couples and HIV-1-unexposed seronegative controls (HUSNs). We found low-frequency HIV-1-specific T cells in both HESNs and HUSNs but show that the response rates were higher over time in the former (P = 0.01). Furthermore, the magnitudes of the HIV-1-specific T cell responses were significantly higher among responding HESNs than among HUSNs over time (P = 0.002). In both groups, responses were mediated by CD4 T cells. The responses were mapped to single peptides, which often corresponded to epitopes restricted by multiple HLA-DR types that have previously been detected in HIV-1-infected patients. HIV-1-specific T cell responses in HUSNs and some HESNs likely represent cross-reactivity to self or foreign non-HIV-1 antigens. The significantly greater T cell responses in HESNs, including in two who were homozygous for CCR5Δ32, demonstrates that HIV-1-specific T cell responses can be induced or augmented by exposure to HIV-1 without infection.
Collapse
Affiliation(s)
- Adam J. Ritchie
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Suzanne L. Campion
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jakub Kopycinski
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Zoe Moodie
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Z. Maggie Wang
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Kruti Pandya
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Stephen Moore
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Michael K. P. Liu
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Simon Brackenridge
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Kristin Kuldanek
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Kenneth Legg
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Myron S. Cohen
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Eric L. Delwart
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Barton F. Haynes
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Sarah Fidler
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Andrew J. McMichael
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Nilu Goonetilleke
- The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom, Statistical Center for HIV/AIDS Research and Prevention (SCHARP), Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, St. Mary's Hospital, Imperial College, London, United Kingdom, Division of Infectious Disease, University of North Carolina, Chapel Hill, North Carolina, Blood Systems Research Institute, San Francisco, California, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
56
|
Delwart E, Bernardin F, Lee TH, Winkelman V, Liu C, Sheppard H, Liu A, Greenblatt R, Anastos K, DeHovitz J, Nowicki M, Cohen M, Golub ET, Barbour J, Buchbinder S, Busch MP. Absence of reproducibly detectable low-level HIV viremia in highly exposed seronegative men and women. AIDS 2011; 25:619-23. [PMID: 21297421 DOI: 10.1097/qad.0b013e3283440269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Transient HIV infections have been invoked to account for the cellular immune responses detected in highly virus-exposed individuals who have remained HIV-seronegative. We tested for very low levels of HIV RNA in 524 seronegative plasma samples from 311 highly exposed women and men from three longitudinal HIV cohorts. DESIGN Two thousand and seventy-three transcription-mediated amplification (TMA) HIV RNA tests were performed for an average of 3.95 TMA assays per plasma sample. Quadruplicate TMA assays, analyzing a total of 2 ml of plasma, provided an estimated sensitivity of 3.5 HIV RNA copies/ml. RESULTS Four samples from individuals who did not seroconvert within the following 6 months were positive for HIV RNA. For one sample, human polymorphism DNA analysis indicated a sample mix-up. Borderline HIV RNA detection signals were detected for the other three positive samples but further replicate TMA testing yielded no positive results. Nested PCR assays (n = 254) for HIV proviral DNA in peripheral blood mononuclear cells (PBMCs) from these three individuals were negative. CONCLUSION Transient viremia was not reproducibly detected in highly HIV-exposed seronegative men and women. If transient infections do occur, plasma HIV RNA levels may remain below the detection limits of the sensitive assay used here, be of very short duration, or viral replication may be restricted to mucosal surfaces or their draining lymphoid tissues.
Collapse
|
57
|
Koup RA, Graham BS, Douek DC. The quest for a T cell-based immune correlate of protection against HIV: a story of trials and errors. Nat Rev Immunol 2010; 11:65-70. [PMID: 21164527 DOI: 10.1038/nri2890] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Even before the partial success of a preventive HIV vaccine in a recent Phase III clinical trial, there had been an active research effort to determine one or more immune correlates of protection for HIV infection. This effort has been hampered by the lack of natural protective immunity against HIV. As a result, most of the studies have focused on long-term non-progressive infection or other clinical situations, none of which fully recapitulates protective immunity against HIV. Although this effort has been successful in defining characteristics of T cells in acute and non-progressive HIV infection, and has therefore greatly expanded our knowledge of the immunopathogenesis of AIDS, its success in defining immune correlates of protection is less clear. In this Opinion article we offer a perspective on how successful this effort has been in defining immune correlates of protection that have been, or will be, of use in the development of an HIV vaccine. Our view is that investing in an iterative approach to human vaccine efficacy trials of sufficient size and sampling frequency will improve the likelihood that an immune correlate of vaccine protection will be defined.
Collapse
Affiliation(s)
- Richard A Koup
- Richard A. Koup, Barney S. Graham and Daniel C. Douek are at the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-3017, USA
| | | | | |
Collapse
|
58
|
Songok EM, Osero B, McKinnon L, Rono MK, Apidi W, Matey EJ, Meyers AFA, Luo M, Kimani J, Wachihi C, Ball BT, Plummer FA, Mpoke S. CD26/dipeptidyl peptidase IV (CD26/DPPIV) is highly expressed in peripheral blood of HIV-1 exposed uninfected female sex workers. Virol J 2010; 7:343. [PMID: 21108831 PMCID: PMC3009705 DOI: 10.1186/1743-422x-7-343] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 11/25/2010] [Indexed: 11/22/2022] Open
Abstract
Background Design of effective vaccines against the human immunodeficiency virus (HIV-1) continues to present formidable challenges. However, individuals who are exposed HIV-1 but do not get infected may reveal correlates of protection that may inform on effective vaccine design. A preliminary gene expression analysis of HIV resistant female sex workers (HIV-R) suggested a high expression CD26/DPPIV gene. Previous studies have indicated an anti-HIV effect of high CD26/DPPIV expressing cells in vitro. Similarly, high CD26/DPPIV protein levels in vivo have been shown to be a risk factor for type 2 diabetes. We carried out a study to confirm if the high CD26/DPPIV gene expression among the HIV-R were concordant with high blood protein levels and its correlation with clinical type 2 diabetes and other perturbations in the insulin signaling pathway. Results A quantitative CD26/DPPIV plasma analysis from 100 HIV-R, 100 HIV infected (HIV +) and 100 HIV negative controls (HIV Neg) showed a significantly elevated CD26/DPPIV concentration among the HIV-R group (mean 1315 ng/ml) than the HIV Neg (910 ng/ml) and HIV + (870 ng/ml, p < 0.001). Similarly a FACs analysis of cell associated DPPIV (CD26) revealed a higher CD26/DPPIV expression on CD4+ T-cells derived from HIV-R than from the HIV+ (90.30% vs 80.90 p = 0.002) and HIV Neg controls (90.30% vs 82.30 p < 0.001) respectively. A further comparison of the mean fluorescent intensity (MFI) of CD26/DPPIV expression showed a higher DPP4 MFI on HIV-R CD4+ T cells (median 118 vs 91 for HIV-Neg, p = 0.0003). An evaluation for hyperglycemia, did not confirm Type 2 diabetes but an impaired fasting glucose condition (5.775 mmol/L). A follow-up quantitative PCR analysis of the insulin signaling pathway genes showed a down expression of NFκB, a central mediator of the immune response and activator of HIV-1 transcription. Conclusion HIV resistant sex workers have a high expression of CD26/DPPIV in tandem with lowered immune activation markers. This may suggest a novel role for CD26/DPPIV in protection against HIV infection in vivo.
Collapse
Affiliation(s)
- Elijah M Songok
- Centre For Virus Research, Mbagathi Road Kenya Medical Research Institute, Nairobi, Kenya.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Martinon F, Kaldma K, Sikut R, Culina S, Romain G, Tuomela M, Adojaan M, Männik A, Toots U, Kivisild T, Morin J, Brochard P, Delache B, Tripiciano A, Ensoli F, Stanescu I, Le Grand R, Ustav M. Persistent immune responses induced by a human immunodeficiency virus DNA vaccine delivered in association with electroporation in the skin of nonhuman primates. Hum Gene Ther 2010; 20:1291-307. [PMID: 19627235 DOI: 10.1089/hum.2009.044] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Strategies to improve vaccine efficacy are still required, especially in the case of chronic infections, including human immunodeficiency virus (HIV). DNA vaccines have potential advantages over conventional vaccines; however, low immunological efficacy has been demonstrated in many experiments involving large animals and in clinical trials. To improve the immunogenicity of DNA vaccines, we have designed a plasmid vector exploiting the binding capacity of the bovine papillomavirus E2 protein and we have used electroporation (EP) to increase DNA uptake after intradermal inoculation. We demonstrated, in nonhuman primates (NHPs), efficient induction of anti-HIV immunity with an improved DNA vaccine vector encoding an artificial fusion protein, consisting of several proteins and selected epitopes from HIV-1. We show that a DNA vaccine delivery method combining intradermal injection and noninvasive EP dramatically increased expression of the vaccine antigen selectively in the epidermis, and our observations strongly suggest the involvement of Langerhans cells in the strength and quality of the anti-HIV immune response. Although the humoral responses to the vaccine were transient, the cellular responses were exceptionally robust and persisted, at high levels, more than 2 years after the last vaccine boost. The immune responses were characterized by the induction of significant proportions of T cells producing both interferon-gamma and interleukin-2 cytokines, in both subpopulations, CD4(+) and CD8(+). This strategy is an attractive approach for vaccination in humans because of its high efficacy and the possible use of newly developed devices for EP.
Collapse
Affiliation(s)
- Frédéric Martinon
- Division of Immunovirology, Life Sciences Program (DSV), Institute for Emerging Diseases and Innovative Therapies, Atomic Energy Commission (CEA), Fontenay aux Roses, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
van der Kuyl AC, Kozaczynska K, Ariën KK, Gali Y, Balázs VR, Dekker SJ, Zorgdrager F, Vanham G, Berkhout B, Cornelissen M. Analysis of infectious virus clones from two HIV-1 superinfection cases suggests that the primary strains have lower fitness. Retrovirology 2010; 7:60. [PMID: 20646276 PMCID: PMC2918528 DOI: 10.1186/1742-4690-7-60] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 07/20/2010] [Indexed: 11/30/2022] Open
Abstract
Background Two HIV-1 positive patients, L and P, participating in the Amsterdam Cohort studies acquired an HIV-1 superinfection within half a year from their primary HIV-1 infection (Jurriaans et al., JAIDS 2008, 47:69-73). The aim of this study was to compare the replicative fitness of the primary and superinfecting HIV-1 strains of both patients. The use of isolate-specific primer sets indicated that the primary and secondary strains co-exist in plasma at all time points after the moment of superinfection. Results Biological HIV-1 clones were derived from peripheral blood CD4 + T cells at different time point, and identified as the primary or secondary virus through sequence analysis. Replication competition assays were performed with selected virus pairs in PHA/IL-2 activated peripheral blood mononuclear cells (PBMC's) and analyzed with the Heteroduplex Tracking Assay (HTA) and isolate-specific PCR amplification. In both cases, we found a replicative advantage of the secondary HIV-1 strain over the primary virus. Full-length HIV-1 genomes were sequenced to find possible explanations for the difference in replication capacity. Mutations that could negatively affect viral replication were identified in the primary infecting strains. In patient L, the primary strain has two insertions in the LTR promoter, combined with a mutation in the tat gene that has been associated with decreased replication capacity. The primary HIV-1 strain isolated from patient P has two mutations in the LTR that have been associated with a reduced replication rate. In a luciferase assay, only the LTR from the primary virus of patient P had lower transcriptional activity compared with the superinfecting virus. Conclusions These preliminary findings suggest the interesting scenario that superinfection occurs preferentially in patients infected with a relatively attenuated HIV-1 isolate.
Collapse
Affiliation(s)
- Antoinette C van der Kuyl
- Laboratory of Experimental Virology, Department of Medical Microbiology, Centre for Infection and Immunity Amsterdam (CINIMA), Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Ji H, Ball TB, Ao Z, Kimani J, Yao X, Plummer FA. Reduced HIV-1 long terminal repeat transcription in subjects with protective interferon regulatory factor-1 genotype: a potential mechanism mediating resistance to infection by HIV-1. ACTA ACUST UNITED AC 2010; 42:389-94. [PMID: 20100115 DOI: 10.3109/00365540903496536] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We previously described the polymorphism in the interferon regulatory factor-1 (IRF-1) gene as a novel correlate of resistance to HIV-1 infection in a Kenyan female sex worker cohort. However, the underlying mechanisms likely mediating this association remained to be elucidated. The initiation of HIV-1 long terminal repeat (LTR) transcription in peripheral blood mononuclear cells (PBMCs) from subjects with different IRF-1 haplotypes, representing protective, intermediate and the least protective IRF-1 allele combinations, were investigated here. A single-cycle pseudovirus construct expressing vesicular stomatitis virus envelop G-protein (VSV-G) and having an HIV-1 pNL4.3 backbone with luciferase insert was used to infect PBMCs with different IRF-1 haplotypes. The efficiency of early HIV-1 LTR transcription was monitored using a luciferase assay. IRF-1 protein levels induced by the infection were measured by quantitative Western blot. Our results showed that PBMCs with the protective IRF-1 genotype demonstrated significantly lower HIV-1 LTR transcription during the initial stages of infection compared to PBMCs with other haplotypes, which correlated with the kinetics of IRF-1 responsiveness to HIV-1 infection in the cells. It suggests that IRF-1 genotypes alter the efficiency of early HIV-1 LTR transcription, likely via modulating expression of IRF-1. This may represent one mechanism mediating the association between IRF-1 polymorphisms and resistance to HIV-1 infection.
Collapse
Affiliation(s)
- Hezhao Ji
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.
| | | | | | | | | | | |
Collapse
|
62
|
HIV viral set point and host immune control in individuals with HIV-specific CD8+ T-cell responses prior to HIV acquisition. AIDS 2010; 24:1449-54. [PMID: 20549840 DOI: 10.1097/qad.0b013e3283391d40] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Vaccine-induced CD8(+) T-cell responses in primates have been associated with a reduced simian immunodeficiency virus plasma viral load and enhanced T-cell responses, but cellular vaccines have shown limited success in human trials. We previously described HIV-specific T-cell responses in two groups of highly exposed, persistently seronegative Kenyan female sex workers, and a subset of these participants have subsequently acquired HIV. We examined the impact of pre-existing CD8(+) T-cell responses on post-acquisition outcomes. DESIGN AND METHODS HIV-specific CD8(+) T-cell responses had been examined in highly exposed, persistently seronegative participants from the Pumwani and Kibera cohorts, using a combination of virus-specific lysis, proliferation, interferon-gamma production, or all. Plasma viral load set point and HIV-specific T-cell proliferation and cytokine production were now examined post hoc by blinded investigators in the subset of participants who acquired HIV. RESULTS Pre-acquisition cellular immune assays and post-infection viral load were available for 46 participants, and HIV-specific CD8(+) T-cell responses had been detected in 25 of 46 (54%) participants. Pre-acquisition CD8(+) T-cell responses were associated with a lower post-acquisition HIV viral load set point in both cohorts (pooled analysis, 3.1 vs. 4.1 log(10) RNA copies/ml; P=0.0002) and with enhanced post-acquisition HIV-specific CD8(+) T-cell proliferation (3.8 vs. 1.0%, P=0.03), but with a trend to reduced post-acquisition CD8(+) T-cell interferon-gamma responses. CONCLUSION HIV-specific CD8(+) T-cell responses prior to HIV acquisition were associated with a lower HIV viral load and an altered functional profile of post-acquisition CD8(+) T-cell responses.
Collapse
|
63
|
Innovative bioinformatic approaches for developing peptide-based vaccines against hypervariable viruses. Immunol Cell Biol 2010; 89:81-9. [PMID: 20458336 DOI: 10.1038/icb.2010.65] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The application of the fields of pharmacogenomics and pharmacogenetics to vaccine design has been recently labeled 'vaccinomics'. This newly named area of vaccine research, heavily intertwined with bioinformatics, seems to be leading the charge in developing novel vaccines for currently unmet medical needs against hypervariable viruses such as human immunodeficiency virus (HIV), hepatitis C and emerging avian and swine influenza. Some of the more recent bioinformatic approaches in the area of vaccine research include the use of epitope determination and prediction algorithms for exploring the use of peptide epitopes as vaccine immunogens. This paper briefly discusses and explores some current uses of bioinformatics in vaccine design toward the pursuit of peptide vaccines for hypervariable viruses. The various informatics and vaccine design strategies attempted by other groups toward hypervariable viruses will also be briefly examined, along with the strategy used by our group in the design and synthesis of peptide immunogens for candidate HIV and influenza vaccines.
Collapse
|
64
|
Davydov DM, Stewart R, Ritchie K, Chaudieu I. Resilience and mental health. Clin Psychol Rev 2010; 30:479-95. [PMID: 20395025 DOI: 10.1016/j.cpr.2010.03.003] [Citation(s) in RCA: 578] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 03/09/2010] [Accepted: 03/17/2010] [Indexed: 01/01/2023]
Abstract
The relationship between disease and good health has received relatively little attention in mental health. Resilience can be viewed as a defence mechanism, which enables people to thrive in the face of adversity and improving resilience may be an important target for treatment and prophylaxis. Though resilience is a widely-used concept, studies vary substantially in their definition, and measurement. Above all, there is no common underlying theoretical construct to this very heterogeneous research which makes the evaluation and comparison of findings extremely difficult. Furthermore, the varying multi-disciplinary approaches preclude meta-analysis, so that clarification of research in this area must proceed firstly by conceptual unification. We attempt to collate and classify the available research around a multi-level biopsychosocial model, theoretically and semiotically comparable to that used in describing the complex chain of events related to host resistance in infectious disease. Using this underlying construct we attempt to reorganize current knowledge around a unitary concept in order to clarify and indicate potential intervention points for increasing resilience and positive mental health.
Collapse
|
65
|
Shankarkumar U, Pawar A, Ghosh K, Bajpai S, Pazare A. Human leucocyte antigen class II DRB1 and DQB1 associations in human immunodeficiency virus-infected patients of Mumbai, India. Int J Immunogenet 2010; 37:199-204. [PMID: 20345872 DOI: 10.1111/j.1744-313x.2010.00911.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The pathogenesis of human immunodeficiency virus (HIV) infection clearly involves immunoregulatory host factors and products of major histocompatibility complex class II genes, which present antigenic peptides to the T-cell receptor on CD4+ cells, which in turn increase the production of specific antibodies and cytotoxic T lymphocytes. The main objective of this study was to determine the associations of human leucocyte antigen (HLA) DRB1 and DQB1 alleles and their haplotypes in 210 HIV-1-infected patients and compare them with 129 healthy normal individuals with same ethnic background. The HLA DRB1 and DQB1 alleles were genotyped using polymerase chain reaction product and sequence-specific probes for reverse line hybridization, analysed with the Invitrogen Dynal PMP software. Our results revealed a highly significant increase of HLA DRB1*0902 [odds ratio (OR) = 17.12; P = 0.004], DQB1*030103 (OR = 53.53; P = 4.61E-07) and DQB1*050201 (OR = 16.26; P = 0.0002) alleles while in contrast highly significant decrease in frequency of HLA DQB1*030101 (OR = 0.36; P = 0.0002), DQB1*050301 (OR = 0.22; P < 0.0001) and DQB1*060101 (OR = 0.43; P < 0.0001) among the HIV-1-infected patients when compared with the controls. The haplotype DRB1*0902-DQB1*030103 (OR = 10.65; P = 0.06) was significantly increased in HIV1 patients, while haplotypes DRB1*150101-DQB1*060101 (OR = 0.386, P < 0.0001), DRB1*030101-DQB1*020101 (OR = 0.197, P = 0.004) and DRB1*070101-DQB1*0202 (OR = 0.167, P = 0.001) were significantly decreased. Our results indicate clearly that there are HLA class II alleles involved in the susceptibility to and protection from HIV-1 infection in our study group and further they vary in different ethnic groups reported in literature.
Collapse
Affiliation(s)
- U Shankarkumar
- National Institute of Immunohaematology, ICMR, KEM Hospital, Parel, Mumbai, India.
| | | | | | | | | |
Collapse
|
66
|
Mölder T, Adojaan M, Kaldma K, Ustav M, Sikut R. Elicitation of broad CTL response against HIV-1 by the DNA vaccine encoding artificial multi-component fusion protein MultiHIV--study in domestic pigs. Vaccine 2009; 28:293-8. [PMID: 19879232 DOI: 10.1016/j.vaccine.2009.10.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 10/08/2009] [Accepted: 10/12/2009] [Indexed: 10/20/2022]
Abstract
Broad CTL response against HIV-1 is one factor that helps to control the viral replication. We have constructed a DNA vaccine that encodes a large artificial fusion protein (MultiHIV) and shown it to be immunogenic in mice, swine and macaques. Inbred mice revealed CTL response only against two epitopes due to limited MHC class I variability. To assess the quality of the CTL response we addressed this question in domestic swine. Number of presented epitopes varied between 7 and 14 among the five selected animals. Epitopes detected in swine are localised in the same antigenic regions recognised in humans. This can be explained by the fact that swine MHC-I (SLA-I) complex is remarkably similar to human HLA-I. These results also indicate that immunogenicity profile of vaccines in domestic swine may predict the outcome of human immunisation.
Collapse
|
67
|
Kersh EN, Luo W, Adams DR, Srinivasan P, Smith JM, Promadej-Lanier N, Ellenberger D, Garcia-Lerma JG, Butera S, Otten R. Repeated rectal SHIVSF162P3 exposures do not consistently induce sustained T cell responses prior to systemic infection in the repeat-low dose preclinical macaque model. AIDS Res Hum Retroviruses 2009; 25:905-17. [PMID: 19689194 DOI: 10.1089/aid.2008.0287] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The macaque model of repeated SHIV exposures is increasingly used as a preclinical tool to evaluate biomedical HIV intervention strategies. It is unclear whether multiple virus exposures induce immune responses in macaques, as documented in uninfected individuals repeatedly exposed to HIV. We here address whether repeated, rectal SHIV(SF162P3) exposures lead to systemic T cell activation in 12 rhesus macaques, and whether this is associated with increased infection resistance. Eight macaques became systemically infected after 2-7 exposures, three macaques were less susceptible (infection after 10-12 exposures), and one macaque remained uninfected after 14 exposures. PBMCs were retrospectively monitored for increases in T cell activation by analyzing the proportion of CD8(+) T cells, recently activated or proliferated T cells (markers CD38, Ki67), a marker for cytotoxicity (granzyme B), or T cell-produced plasma cytokines (IFN-gamma, RANTES, IL-2). Repeated virus exposures did not induce sustained, potent, or diverse T cell responses prior to systemic infection. Some changes occurred in the analyzed parameters during repeated virus exposures, but similar T cell activities were also observed in five SHIV-unexposed control macaques. Thus, we found no evidence that delayed infection or resistance to infection was associated with systemic, long-lasting, protective T cell responses to repeated rectal virus exposures. Our results provide further insights into the repeat exposure macaque model. We find that this model can be used for testing biomedical prevention strategies without concern of eliciting a systemic vaccination effect.
Collapse
Affiliation(s)
- Ellen N. Kersh
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Wei Luo
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Debra R. Adams
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Priya Srinivasan
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - James M. Smith
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Nattawan Promadej-Lanier
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Dennis Ellenberger
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - J. Gerardo Garcia-Lerma
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Salvatore Butera
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Ron Otten
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| |
Collapse
|
68
|
Preinfection human immunodeficiency virus (HIV)-specific cytotoxic T lymphocytes failed to prevent HIV type 1 infection from strains genetically unrelated to viruses in long-term exposed partners. J Virol 2009; 83:10821-9. [PMID: 19706711 DOI: 10.1128/jvi.00839-09] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms underlying potential altered susceptibility to human immunodeficiency virus type 1 (HIV-1) infection in highly exposed seronegative (ES) individuals and the later clinical consequences of breakthrough infection can provide insight into strategies to control HIV-1 with an effective vaccine. From our Seattle ES cohort, we identified one individual (LSC63) who seroconverted after over 2 years of repeated unprotected sexual contact with his HIV-1-infected partner (P63) and other sexual partners of unknown HIV-1 serostatus. The HIV-1 variants infecting LSC63 were genetically unrelated to those sequenced from P63. This may not be surprising, since viral load measurements in P63 were repeatedly below 50 copies/ml, making him an unlikely transmitter. However, broad HIV-1-specific cytotoxic T-lymphocyte (CTL) responses were detected in LSC63 before seroconversion. Compared to those detected after seroconversion, these responses were of lower magnitude and half of them targeted different regions of the viral proteome. Strong HLA-B27-restricted CTLs, which have been associated with disease control, were detected in LSC63 after but not before seroconversion. Furthermore, for the majority of the protein-coding regions of the HIV-1 variants in LSC63 (except gp41, nef, and the 3' half of pol), the genetic distances between the infecting viruses and the viruses to which he was exposed through P63 (termed the exposed virus) were comparable to the distances between random subtype B HIV-1 sequences and the exposed viruses. These results suggest that broad preinfection immune responses were not able to prevent the acquisition of HIV-1 infection in LSC63, even though the infecting viruses were not particularly distant from the viruses that may have elicited these responses.
Collapse
|
69
|
Bell SK, Rosenberg ES. Case records of the Massachusetts General Hospital. Case 11-2009. A 47-year-old man with fever, headache, rash, and vomiting. N Engl J Med 2009; 360:1540-8. [PMID: 19357410 DOI: 10.1056/nejmcpc0810840] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Sigall K Bell
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center and the Department of Medicine, Harvard Medical School, USA
| | | |
Collapse
|
70
|
Abstract
More than 25 years after the recognition of AIDS and the isolation of the causative agent, human immunodeficiency virus (HIV), we have been unable to develop a vaccine to protect against infection. The major obstacle to development of a vaccine has been the absence of naturally acquired protective immunity, which is characteristic of most infectious agents. We and others, however, have identified individuals who appear to be resistant to infection. Using a combination of epidemiology, molecular biology, and genetics, we hypothesize that these individuals are able to resist infection by clearing low doses of HIV from their systems. We further hypothesize that they are able to clear the virus through a highly efficient system of processing and presentation of HIV epitopes (antigens) to CD8+ cytotoxic cells, which activate them to remove virally infected cells. Subsequent studies have lent support to this hypothesis.
Collapse
Affiliation(s)
- Roger Detels
- School of Public Health, Department of Epidemiology, University of California-Los Angeles, 650 Charles Young Drive, Los Angeles, CA 90085-1772, USA.
| |
Collapse
|
71
|
Co MDT, Kilpatrick ED, Rothman AL. Dynamics of the CD8 T-cell response following yellow fever virus 17D immunization. Immunology 2009; 128:e718-27. [PMID: 19740333 DOI: 10.1111/j.1365-2567.2009.03070.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Management of yellow fever is focused on the prevention of illness by the use of the yellow fever virus (YFV) 17D vaccine. The role of neutralizing antibodies in protection is generally accepted with YFV-specific T cells likely contributing to the control of viral replication. We studied CD8(+) T-cell responses to four defined human leucocyte antigen-B35-restricted epitopes in YFV vaccine recipients as a model of the kinetics of cytotoxic T-lymphocyte responses to an acute human viral infection. Multiple features of these epitope-specific responses were analysed after vaccination including magnitude, cytokine production, phenotype and T-cell receptor repertoire. Peak peptide-specific interferon-gamma (IFN-gamma) responses of almost 1% of CD8(+) T cells were seen as early as 2 weeks post-vaccination; however, dominant responses varied between donors. Peptide-specific responses were still detectable at 54 months post-vaccination. Tetramer-positive cells, at high frequencies, were detected as early as 7-9 days, before detectable IFN-gamma-producing cells, suggesting a defect in the functional capacity of some antigen-specific cells early post-vaccination. The predominant memory phenotype of the tetramer-positive population was a differentiated effector (CD45RA(+) CCR7(-) CD62L(-)) phenotype. The T-cell receptor Vbeta analysis revealed a diverse oligoclonal repertoire in tetramer-positive T-cell populations in two individuals. These characteristics of the YFV-specific T-cell response could contribute to vaccine effectiveness.
Collapse
Affiliation(s)
- Mary Dawn T Co
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | |
Collapse
|
72
|
|
73
|
Abstract
Small animal models in which in vivo HIV-1 infection, pathogenesis, and immune responses can be studied would permit both basic research on the biology of the disease, as well as a system to rapidly screen developmental therapeutics and/or vaccines. To date, the most widely-used models have been the severe combined immunodeficient (SCID)-hu (also known as the thy/liv SCID-hu) and the huPBL-SCID mouse models. Recently three new models have emerged, i.e., the intrasplenic huPBL/SPL-SCID model, the NOD/SCID/IL2Rgamma(null) mouse model, and the Rag2(-/-)gamma(c) (-/-) mouse model. Details on the construction, maintenance and HIV-1 infection of these models are discussed.
Collapse
Affiliation(s)
- Aviva Joseph
- Department of Microbiology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | |
Collapse
|
74
|
Blondelle SE, Moya-Castro R, Osawa K, Schroder K, Wilson DB. Immunogenically optimized peptides derived from natural mutants of HIV CTL epitopes and peptide combinatorial libraries. Biopolymers 2008; 90:683-94. [PMID: 18481808 DOI: 10.1002/bip.21020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Two strategies were aimed at identifying immunogenically optimized peptides for the potential use in the formulation of an effective prophylactic or therapeutic HIV-1 vaccine. Three CTL epitopes were investigated: Gag p24(19-27) TV9, Gag p17(77-85) SL9, and RT(309-317) IV9. The first strategy derives from the hypothesis that a number of rare mutant CTL epitopes of HIV-1 may be more immunogenic than the common ones. As such, these rare mutant sequences might be highly effective in generating cross reactive anti-HIV-1 CTL responses against a range of mutant sequences. As anticipated, several rare mutant peptide sequences were identified that generated strong CTL responses against both the consensus sequences and several naturally occurring mutants in human PBL cultures primed ex vivo and in HLA-A2 transgenic mice immunized in vivo. Finally, to reach beyond the sequence diversity of the "natural" library of mutated sequences, a synthetic combinatorial peptide library was screened with a TV9 specific T-cell line; this resulted in the identification of an immunogenically optimized mimic peptide sequence that provoked highly effective CTL immune responses against TV9 and mutants. Sequence homologies between the natural mutants and synthetic mimic may provide insight into key contact positions in the MHC/TCR/peptide complex.
Collapse
Affiliation(s)
- Sylvie E Blondelle
- Mixture Sciences, Inc., 3550 General Atomics Ct, San Diego, CA 92121, USA.
| | | | | | | | | |
Collapse
|
75
|
Huang S, Dunkley-Thompson J, Tang Y, Macklin EA, Steel-Duncan J, Singh-Minott I, Ryland EG, Smikle M, Walker BD, Christie CD, Feeney ME. Deficiency of HIV-Gag-specific T cells in early childhood correlates with poor viral containment. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:8103-11. [PMID: 19018003 PMCID: PMC2714370 DOI: 10.4049/jimmunol.181.11.8103] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Perinatal HIV infection is characterized by a sustained high-level viremia and a high risk of rapid progression to AIDS, indicating a failure of immunologic containment of the virus. We hypothesized that age-related differences in the specificity or function of HIV-specific T cells may influence HIV RNA levels and clinical outcome following perinatal infection. In this study, we defined the HIV epitopes targeted by 76 pediatric subjects (47 HIV infected and 29 HIV exposed, but uninfected), and assessed the ability of HIV-specific CD8 and CD4 T cells to degranulate and produce IFN-gamma, TNF-alpha, and IL-2. No responses were detected among HIV-uninfected infants, whereas responses among infected subjects increased in magnitude and breadth with age. Gag-specific responses were uncommon during early infancy, and their frequency was significantly lower among children younger than 24 mo old (p = 0.014). Importantly, Gag responders exhibited significantly lower HIV RNA levels than nonresponders (log viral load 5.8 vs 5.0; p = 0.005). Both the total and Gag-specific T cell frequency correlated inversely with viral load after correction for age, whereas no relationship with targeting of other viral proteins was observed. Functional assessment of HIV-specific T cells by multiparameter flow cytometry revealed that polyfunctional CD8 cells were less prevalent in children before 24 mo of age, and that HIV-specific CD4 cell responses were of universally low frequency among antiretroviral-naive children and absent in young infants. These cross-sectional data suggest that qualitative differences in the CD8 response, combined with a deficiency of HIV-specific CD4 cells, may contribute to the inability of young infants to limit replication of HIV.
Collapse
Affiliation(s)
- SiHong Huang
- Partners AIDS Research Center and Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115
- Children's Hospital Boston, Boston, MA 02115
- Clinical Investigator Training Program: Harvard/MIT Health Sciences and Technology - Beth Israel Deaconess Medical Center, in collaboration with Pfizer Inc. and Merck Co., Boston, MA 02114
| | - Jacqueline Dunkley-Thompson
- Kingston Perinatal AIDS Program and Department of Obstetrics, Gynecology and Pediatrics, University of the West Indies, Kingston, Jamaica
| | - YanHua Tang
- Partners AIDS Research Center and Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115
| | - Eric A. Macklin
- MGH Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114
| | - Julianne Steel-Duncan
- Kingston Perinatal AIDS Program and Department of Obstetrics, Gynecology and Pediatrics, University of the West Indies, Kingston, Jamaica
| | - Indira Singh-Minott
- Kingston Perinatal AIDS Program and Department of Obstetrics, Gynecology and Pediatrics, University of the West Indies, Kingston, Jamaica
| | - Elizabeth G. Ryland
- Partners AIDS Research Center and Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115
| | - Monica Smikle
- Kingston Perinatal AIDS Program and Department of Obstetrics, Gynecology and Pediatrics, University of the West Indies, Kingston, Jamaica
| | - Bruce D. Walker
- Partners AIDS Research Center and Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115
- Howard Hughes Medical Institute, Chev Chase, MD 20815
| | - Celia D.C. Christie
- Kingston Perinatal AIDS Program and Department of Obstetrics, Gynecology and Pediatrics, University of the West Indies, Kingston, Jamaica
| | - Margaret E. Feeney
- Partners AIDS Research Center and Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115
- Children's Hospital Boston, Boston, MA 02115
| |
Collapse
|
76
|
Klinman D. ELISPOT Assay to Detect Cytokine‐Secreting Murine and Human Cells. ACTA ACUST UNITED AC 2008; Chapter 6:6.19.1-6.19.9. [DOI: 10.1002/0471142735.im0619s83] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Dennis Klinman
- Cancer and Inflammation Program, National Cancer Institute Frederick Maryland
| |
Collapse
|
77
|
Speelmon EC, Livingston-Rosanoff D, Desbien AL, Lee J, Wick WD, Hladik F, McElrath MJ. Impaired viral entry cannot explain reduced CD4+ T cell susceptibility to HIV type 1 in certain highly exposed individuals. AIDS Res Hum Retroviruses 2008; 24:1415-27. [PMID: 19000021 PMCID: PMC2764523 DOI: 10.1089/aid.2007.0256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Rare individuals report repeated unprotected HIV-1 sexual exposures, yet remain seronegative for years. We investigated the possibility that reduced in vitro CD4(+) T cell susceptibility to HIV-1 infection protects such highly exposed seronegative (ES) individuals. Susceptibility to three R5-tropic HIV-1 isolates, regardless of inoculating dose, was remarkably similar between 81 ES and 33 low-risk controls. In 94% (99/105) of donors, we observed a 1.36 log-unit range in HIV-1(JR-CSF) production, with similar results for HIV-1(1192). The median frequency of intracellular Gag(+) T cells after single-round infection was similar in ES (5.2%) and controls (7.2%), p = 0.456. However, in repeated testing, CD4(+) T cells from two controls (6.1%) and four ES (4.9%) exhibited a 10- to 2500-fold reduction in HIV-1 production and required 5- to 12-fold greater HIV-1(1192) and HIV-1(JR-CSF) inocula to establish infection (TCID(50)). Reduced viral entry cannot explain the low producer phenotype; no differences in CCR5 receptor density or beta-chemokine production were observed. In conclusion, we have identified a remarkably narrow range of HIV-1 susceptibility in seronegative donors regardless of risk activity, which can be applied as a benchmark to assess vaccine-induced antiviral effector activities. However, CD4(+) T cells from a subset of individuals demonstrated reduced HIV-1 susceptibility unexplained by impaired entry, lending support to the possibility that cellular restriction of HIV-1 may account for continued seronegativity in some of those having repeated sexual exposure. Identifying the host-virus interactions responsible for diminished in vitro susceptibility may contribute to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Emily C. Speelmon
- Medical Scientist Training Program, University of Washington, Seattle, Washington 98105
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington 98105
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Devon Livingston-Rosanoff
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Anthony L. Desbien
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Jean Lee
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - W. David Wick
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Florian Hladik
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98105
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, Washington 98105
| |
Collapse
|
78
|
Banks HT, Davidian M, Hu S, Kepler GM, Rosenberg ES. Modelling HIV immune response and validation with clinical data. JOURNAL OF BIOLOGICAL DYNAMICS 2008; 2:357-85. [PMID: 19495424 PMCID: PMC2689816 DOI: 10.1080/17513750701813184] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
A system of ordinary differential equations is formulated to describe the pathogenesis of HIV infection, wherein certain features that have been shown to be important by recent experimental research are incorporated in the model. These include the role of CD4+ memory cells that serve as a major reservoir of latently infected cells, a critical role for T-helper cells in the generation of CD8 memory cells capable of efficient recall response, and stimulation by antigens other than HIV. A stability analysis illustrates the capability of this model in admitting multiple locally asymptotically stable (locally a.s.) off-treatment equilibria.We show that this more biologically detailed model can exhibit the phenomenon of transient viremia experienced by some patients on therapy with viral load levels suppressed below the detection limit. We also show that the loss of CD4+ T-cell help in the generation of CD8+ memory cells leads to larger peak values for the viral load during transient viremia. Censored clinical data is used to obtain parameter estimates. We demonstrate that using a reduced set of 16 free parameters, obtained by fixing some parameters at their population averages, the model provides reasonable fits to the patient data and, moreover, that it exhibits good predictive capability. We further show that parameter values obtained for most clinical patients do not admit multiple locally a.s off-treatment equilibria. This suggests that treatment to move from a high viral load equilibrium state to an equilibrium state with a lower (or zero) viral load is not possible for these patients.
Collapse
Affiliation(s)
- H T Banks
- Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC, USA.
| | | | | | | | | |
Collapse
|
79
|
Lee LYH, Ha DLA, Simmons C, de Jong MD, Chau NVV, Schumacher R, Peng YC, McMichael AJ, Farrar JJ, Smith GL, Townsend AR, Askonas BA, Rowland-Jones S, Dong T. Memory T cells established by seasonal human influenza A infection cross-react with avian influenza A (H5N1) in healthy individuals. J Clin Invest 2008; 118:3478-90. [PMID: 18802496 PMCID: PMC2542885 DOI: 10.1172/jci32460] [Citation(s) in RCA: 246] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Accepted: 07/30/2008] [Indexed: 11/17/2022] Open
Abstract
The threat of avian influenza A (H5N1) infection in humans remains a global health concern. Current influenza vaccines stimulate antibody responses against the surface glycoproteins but are ineffective against strains that have undergone significant antigenic variation. An alternative approach is to stimulate pre-existing memory T cells established by seasonal human influenza A infection that could cross-react with H5N1 by targeting highly conserved internal proteins. To determine how common cross-reactive T cells are, we performed a comprehensive ex vivo analysis of cross-reactive CD4+ and CD8+ memory T cell responses to overlapping peptides spanning the full proteome of influenza A/Viet Nam/CL26/2005 (H5N1) and influenza A/New York/232/2004 (H3N2) in healthy individuals from the United Kingdom and Viet Nam. Memory CD4+ and CD8+ T cells isolated from the majority of participants exhibited human influenza-specific responses and showed cross-recognition of at least one H5N1 internal protein. Participant CD4+ and CD8+ T cells recognized multiple synthesized influenza peptides, including peptides from the H5N1 strain. Matrix protein 1 (M1) and nucleoprotein (NP) were the immunodominant targets of cross-recognition. In addition, cross-reactive CD4+ and CD8+ T cells recognized target cells infected with recombinant vaccinia viruses expressing either H5N1 M1 or NP. Thus, vaccine formulas inducing heterosubtypic T cell-mediated immunity may confer broad protection against avian and human influenza A viruses.
Collapse
Affiliation(s)
- Laurel Yong-Hwa Lee
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Do Lien Anh Ha
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Cameron Simmons
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Menno D. de Jong
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Nguyen Van Vinh Chau
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Reto Schumacher
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Yan Chun Peng
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Andrew J. McMichael
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jeremy J. Farrar
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Geoffrey L. Smith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Alain R.M. Townsend
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Brigitte A. Askonas
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Sarah Rowland-Jones
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Tao Dong
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam.
Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom.
Molecular Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
80
|
Mizukoshi E, Eisenbach C, Edlin BR, Newton KP, Raghuraman S, Weiler-Normann C, Tobler LH, Busch MP, Carrington M, McKeating JA, O’Brien TR, Rehermann B. Hepatitis C virus (HCV)-specific immune responses of long-term injection drug users frequently exposed to HCV. J Infect Dis 2008; 198:203-12. [PMID: 18505381 PMCID: PMC2699613 DOI: 10.1086/589510] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Injection drug users (IDUs) who successfully clear hepatitis C virus (HCV) have a reduced risk of developing chronic reinfection, despite their continuing exposure to the virus. To identify immunological correlates for this apparent protection, we studied HCV-specific immune responses in long-term IDUs (duration, >10 years). METHODS HCV-specific T cell responses were assessed in proliferation, enzyme-linked immunospot (ELISPOT), interferon (IFN)-gamma secretion, and cytotoxicity assays, whereas HCV-specific antibodies were assessed in enzyme immunoassays (EIAs), chemiluminescent assays, and in vitro neutralization assays. RESULTS HCV-specific T cell proliferation and IFN-gamma production were more common in nonviremic EIA-positive IDUs (16 [94%] of 17 IDUs) than in viremic EIA-positive IDUs (9 [45%] of 20 IDUs) (P= .003). They were also noted in 16 (62%) of 26 nonviremic EIA-negative IDUs. In contrast, 19 (90%) of 21 viremic IDUs displayed neutralizing antibodies (nAbs), compared with 9 (56%) of 16 nonviremic EIA-positive IDUs (P= .04) and 0 of 24 nonviremic EIA-negative IDUs. Nonviremic IDUs with nAbs were older (P= .0115) than those without nAbs, but these groups did not differ in terms of either injection drug use duration or HCV-specific T cell responses. CONCLUSION The reduced risk of HCV persistence in IDUs previously recovered from HCV infection correlated with T cell responses, and prolonged antigenic stimulation appears to be required to maintain humoral responses.
Collapse
Affiliation(s)
- Eishiro Mizukoshi
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda
| | - Christoph Eisenbach
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda
| | - Brian R. Edlin
- University of California, San Francisco
- Center for the Study of Hepatitis C, Weill Medical College, Cornell University, New York, New York
| | - Kimberly P. Newton
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
| | - Sukanya Raghuraman
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda
| | - Christina Weiler-Normann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda
| | | | - Michael P. Busch
- University of California, San Francisco
- Blood Systems Research Institute, San Francisco, California
| | - Mary Carrington
- Laboratory of Genomic Diversity, Science Applications International Cooperation–Frederick, National Cancer Institute at Frederick, Frederick, Maryland
| | - Jane A. McKeating
- Center for the Study of Hepatitis C, The Rockefeller University, New York, New York
| | - Thomas R. O’Brien
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda
| |
Collapse
|
81
|
van Loggerenberg F, Mlisana K, Williamson C, Auld SC, Morris L, Gray CM, Abdool Karim Q, Grobler A, Barnabas N, Iriogbe I, Abdool Karim SS. Establishing a cohort at high risk of HIV infection in South Africa: challenges and experiences of the CAPRISA 002 acute infection study. PLoS One 2008; 3:e1954. [PMID: 18414658 PMCID: PMC2278382 DOI: 10.1371/journal.pone.0001954] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 02/27/2008] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES To describe the baseline demographic data, clinical characteristics and HIV-incidence rates of a cohort at high risk for HIV infection in South Africa as well as the challenges experienced in establishing and maintaining the cohort. METHODOLOGY/PRINCIPLE FINDINGS Between August 2004 and May 2005 a cohort of HIV-uninfected women was established for the CAPRISA 002 Acute Infection Study, a natural history study of HIV-1 subtype C infection. Volunteers were identified through peer-outreach. The cohort was followed monthly to determine HIV infection rates and clinical presentation of early HIV infection. Risk reduction counselling and male and female condoms were provided. After screening 775 individuals, a cohort of 245 uninfected high-risk women was established. HIV-prevalence at screening was 59.6% (95% CI: 55.9% to 62.8%) posing a challenge in accruing HIV-uninfected women. The majority of women (78.8%) were self-identified as sex-workers with a median of 2 clients per day. Most women (95%) reported more than one casual sexual partner in the previous 3 months (excluding clients) and 58.8% reported condom use in their last sexual encounter. Based on laboratory testing, 62.0% had a sexually transmitted infection at baseline. During 390 person-years of follow-up, 28 infections occurred yielding seroincidence rate of 7.2 (95% CI: 4.5 to 9.8) per 100 person-years. Despite the high mobility of this sex worker cohort retention rate after 2 years was 86.1%. High co-morbidity created challenges for ancillary care provision, both in terms of human and financial resources. CONCLUSIONS/SIGNIFICANCE Challenges experienced were high baseline HIV-prevalence, lower than anticipated HIV-incidence and difficulties retaining participants. Despite challenges, we have successfully accrued this cohort of HIV-uninfected women with favourable retention, enabling us to study the natural history of HIV-1 during acute HIV-infection. Our experiences provide lessons for others establishing similar cohorts, which will be key for advancing the vaccine and prevention research agenda in resource-constrained settings.
Collapse
Affiliation(s)
- Francois van Loggerenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Cohen MS, Hellmann N, Levy JA, DeCock K, Lange J. The spread, treatment, and prevention of HIV-1: evolution of a global pandemic. J Clin Invest 2008; 118:1244-54. [PMID: 18382737 PMCID: PMC2276790 DOI: 10.1172/jci34706] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The most up-to-date estimates demonstrate very heterogeneous spread of HIV-1, and more than 30 million people are now living with HIV-1 infection, most of them in sub-Saharan Africa. The efficiency of transmission of HIV-1 depends primarily on the concentration of the virus in the infectious host. Although treatment with antiviral agents has proven a very effective way to improve the health and survival of infected individuals, as we discuss here, the epidemic will continue to grow unless greatly improved prevention strategies can be developed and implemented. No prophylactic vaccine is on the horizon. However, several behavioral and structural strategies have made a difference--male circumcision provides substantial protection from sexually transmitted diseases, including HIV-1, and the application of antiretroviral agents for prevention holds great promise.
Collapse
Affiliation(s)
- Myron S Cohen
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, USA.
| | | | | | | | | |
Collapse
|
83
|
Azizi A, Anderson DE, Torres JV, Ogrel A, Ghorbani M, Soare C, Sandstrom P, Fournier J, Diaz-Mitoma F. Induction of Broad Cross-Subtype-Specific HIV-1 Immune Responses by a Novel Multivalent HIV-1 Peptide Vaccine in Cynomolgus Macaques. THE JOURNAL OF IMMUNOLOGY 2008; 180:2174-86. [DOI: 10.4049/jimmunol.180.4.2174] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
84
|
An asymmetric model of heterozygote advantage at major histocompatibility complex genes: degenerate pathogen recognition and intersection advantage. Genetics 2008; 178:1473-89. [PMID: 18245836 DOI: 10.1534/genetics.107.082131] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We characterize the function of MHC molecules by the sets of pathogens that they recognize, which we call their "recognition sets." Two features of the MHC-pathogen interaction may be important to the theory of polymorphism construction at MHC loci: First, there may be a large degree of overlap, or degeneracy, among the recognition sets of MHC molecules. Second, when infected with a pathogen, an MHC genotype may have a higher fitness if that pathogen belongs to the overlapping portion, or intersection, of the two recognition sets of the host, when compared with a genotype that contains that pathogen in only one of its recognition sets. We call this benefit "intersection advantage," gamma, and incorporate it, as well as the degree of recognition degeneracy, m, into a model of heterozygote advantage that utilizes a set-theoretic definition of fitness. Counterintuitively, we show that levels of polymorphism are positively related to m and that a high level of recognition degeneracy is necessary for polymorphism at MHC loci under heterozygote advantage. Increasing gamma reduces levels of polymorphism considerably. Hence, if intersection advantage is significant for MHC genotypes, then heterozygote advantage may not explain the very high levels of polymorphism observed at MHC genes.
Collapse
|
85
|
Lentiviral vectors encoding human immunodeficiency virus type 1 (HIV-1)-specific T-cell receptor genes efficiently convert peripheral blood CD8 T lymphocytes into cytotoxic T lymphocytes with potent in vitro and in vivo HIV-1-specific inhibitory activity. J Virol 2008; 82:3078-89. [PMID: 18184707 DOI: 10.1128/jvi.01812-07] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1)-specific CD8 cytotoxic T-lymphocyte (CTL) response plays a critical role in controlling HIV-1 replication. Augmenting this response should enhance control of HIV-1 replication and stabilize or improve the clinical course of the disease. Although cytomegalovirus (CMV) or Epstein-Barr virus (EBV) infection in immunocompromised patients can be treated by adoptive transfer of ex vivo-expanded CMV- or EBV-specific CTLs, adoptive transfer of ex vivo-expanded, autologous HIV-1-specific CTLs had minimal effects on HIV-1 replication, likely a consequence of the inherently compromised qualitative function of HIV-1-specific CTLs derived from HIV-1-infected individuals. We hypothesized that this limitation could be circumvented by using as an alternative source of HIV-1-specific CTLs, autologous peripheral CD8(+) T lymphocytes whose antigen specificity is redirected by transduction with lentiviral vectors encoding HIV-1-specific T-cell receptor (TCR) alpha and beta chains, an approach used successfully in cancer therapy. To efficiently convert peripheral CD8 lymphocytes into HIV-1-specific CTLs that potently suppress in vivo HIV-1 replication, we constructed lentiviral vectors encoding the HIV-1-specific TCR alpha and TCR beta chains cloned from a CTL clone specific for an HIV Gag epitope, SL9, as a single transcript linked with a self-cleaving peptide. We demonstrated that transduction with this lentiviral vector efficiently converted primary human CD8 lymphocytes into HIV-1-specific CTLs with potent in vitro and in vivo HIV-1-specific activity. Using lentiviral vectors encoding an HIV-1-specific TCR to transform peripheral CD8 lymphocytes into HIV-1-specific CTLs with defined specificities represents a new immunotherapeutic approach to augment the HIV-1-specific immunity of infected patients.
Collapse
|
86
|
Perspectives for a protective HIV-1 vaccine. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2008; 56:423-52. [PMID: 18086420 DOI: 10.1016/s1054-3589(07)56014-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
87
|
Persistently HIV-1 seronegative Nairobi sex workers are susceptible to in vitro infection. Can J Infect Dis 2007; 11:259-63. [PMID: 18159299 DOI: 10.1155/2000/390310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/1999] [Accepted: 11/10/1999] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To evaluate whether resistance to HIV-1 infection in a subset of highly exposed sex workers correlates with resistance at the cellular level. DESIGN In vitro evaluation of susceptibility to infection by Kenyan HIV-1 isolates and cellular production of potential mediators of resistance. SETTING Samples were collected in a primary care clinic in Nairobi. PATIENTS Thirteen individuals from a cohort of sex workers with a similar risk of acquiring HIV infection and six unexposed controls. INTERVENTIONS Subjects were provided with appropriate primary care and counselling on the prevention of sexually transmitted diseases. RESULTS No inherent cellular resistance to infection was identified. CD8⁺ cells from a subset of subjects strongly inhibited viral replication. CONCLUSIONS Lack of infection in this cohort was not attributable to factors inherent to CD4⁺ cells. Resistance to HIV infection is likely to be multifactorial, and products of CD8⁺ cells and unique features of mucosal sites probably contribute to this state.
Collapse
|
88
|
Not just sheer luck! Immune correlates of protection against HIV-1 infection. Vaccine 2007; 26:3002-7. [PMID: 18180082 DOI: 10.1016/j.vaccine.2007.11.062] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 11/20/2007] [Accepted: 11/23/2007] [Indexed: 01/21/2023]
Abstract
Susceptibility to HIV infection is widely different among individuals, and it is known that individuals can be identified who are repeatedly exposed to HIV but in whom neither infection nor disease are seen. The possibility that sheer luck is not the only determinant of this phenomenon begun to be considered in 1989 when it was reported that T cell responses to HIV proteins could be detected in antibody-negative sexual partners of known HIV-positive men. In this review, we will summarize the body of knowledge that stemmed from that first observation.
Collapse
|
89
|
Saini M, Hadas E, Volsky DJ, Potash MJ. Vaccine-induced protection from infection of mice by chimeric human immunodeficiency virus type 1, EcoHIV/NL4-3. Vaccine 2007; 25:8660-3. [PMID: 18023943 PMCID: PMC2219693 DOI: 10.1016/j.vaccine.2007.10.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 10/04/2007] [Accepted: 10/08/2007] [Indexed: 11/17/2022]
Abstract
EcoHIV/NL4-3 is a chimeric human immunodeficiency virus type 1 (HIV-1) that can productively infect mice. This study tests the utility of EcoHIV/NL4-3 infection to reveal protective immune responses to an HIV-1 vaccine. Immunocompetent mice were first immunized with VRC 4306 which encodes subtype B consensus sequences of gag, pol, and nef and then were infected by EcoHIV/NL4-3. Anti-Gag antibodies were sampled during immunization and infection. The extent of EcoHIV/NL4-3 infection in spleen cells and peritoneal macrophages was determined by quantitative real-time PCR (QPCR). Although antibody titres were not significantly different in control and vaccinated groups, VRC 4306 immunization induced protective responses that significantly reduced virus burden in both lymphocyte and macrophage compartments. These results indicate that EcoHIV/NL4-3 infection can be controlled by HIV-1 vaccine-induced responses, introducing a small animal model to test vaccine efficacy against HIV-1 infection.
Collapse
Affiliation(s)
- Manisha Saini
- Molecular Virology Division, St. Luke’sRoosevelt Hospital Center, Columbia University Medical Center, 432 West 58 Street, New York, NY 10019, USA
| | - Eran Hadas
- Molecular Virology Division, St. Luke’sRoosevelt Hospital Center, Columbia University Medical Center, 432 West 58 Street, New York, NY 10019, USA
| | - David J. Volsky
- Molecular Virology Division, St. Luke’sRoosevelt Hospital Center, Columbia University Medical Center, 432 West 58 Street, New York, NY 10019, USA
| | - Mary Jane Potash
- Molecular Virology Division, St. Luke’sRoosevelt Hospital Center, Columbia University Medical Center, 432 West 58 Street, New York, NY 10019, USA
| |
Collapse
|
90
|
Improved protection against simian immunodeficiency virus mucosal challenge in macaques primed with a DNA vaccine and boosted with the recombinant modified vaccinia virus Ankara and recombinant Semliki Forest virus. Vaccine 2007; 26:532-45. [PMID: 18093703 DOI: 10.1016/j.vaccine.2007.11.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 11/04/2007] [Accepted: 11/11/2007] [Indexed: 12/25/2022]
Abstract
Using the experimental infection of cynomolgus macaques with simian immunodeficiency virus (SIV) as a model of human immunodeficiency virus infection in humans, we studied the immunogenicity and protective efficacy of a vaccine strategy combining DNA, the modified recombinant vaccinia virus strain Ankara (MVA) and Semliki Forest virus (SFV) expressing gag, pol, env, tat, rev and nef from SIV. Although this immunization strategy induced moderate immune responses, the control of pathogenic SIVmac251 infection following mucosal challenge was clearly improved by vaccination. The viral load in vaccinated animals was reduced by 2 logs during the acute phase of infection and, in five of the six macaques, viral load fell below the detection limit at set point. No correlates of immune protection were identified, but SIV-specific T-cell responses were detected earlier in vaccinated animals than in controls. These results highlight the power of live attenuated virus vectors for vaccination strategies.
Collapse
|
91
|
Huang X, Liu L, Ren L, Qiu C, Wan Y, Xu J. Mucosal priming with replicative Tiantan vaccinia and systemic boosting with DNA vaccine raised strong mucosal and systemic HIV-specific immune responses. Vaccine 2007; 25:8874-84. [DOI: 10.1016/j.vaccine.2007.08.066] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 08/21/2007] [Accepted: 08/27/2007] [Indexed: 10/22/2022]
|
92
|
Lisziewicz J, Calarota SA, Lori F. The potential of topical DNA vaccines adjuvanted by cytokines. Expert Opin Biol Ther 2007; 7:1563-74. [PMID: 17916048 DOI: 10.1517/14712598.7.10.1563] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
To improve the efficacy of DNA immunization epidermal Langerhans cells are attractive targets to deliver antigen-encoding plasmid DNA. Topical vaccination with naked plasmid DNA has been shown to induce immune responses, and their potency might be improved by chemical and physical methods aimed to enhance the efficiency of plasmid DNA delivery into the skin. Cytokines have also been evaluated as adjuvants for DNA vaccines because they influence the host immune response. This review focuses on the action of several cytokines tested as molecular adjuvants for DNA vaccines and the combination of them with the DermaVir Patch vaccine. DermaVir vaccine, topically administered under a patch, consists of a plasmid DNA that is chemically formulated into a nanoparticle to support vaccine delivery into epidermal Langerhans cells and to induce antigen-specific memory T cells.
Collapse
|
93
|
Létourneau S, Im EJ, Mashishi T, Brereton C, Bridgeman A, Yang H, Dorrell L, Dong T, Korber B, McMichael AJ, Hanke T. Design and pre-clinical evaluation of a universal HIV-1 vaccine. PLoS One 2007; 2:e984. [PMID: 17912361 PMCID: PMC1991584 DOI: 10.1371/journal.pone.0000984] [Citation(s) in RCA: 239] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 09/13/2007] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND One of the big roadblocks in development of HIV-1/AIDS vaccines is the enormous diversity of HIV-1, which could limit the value of any HIV-1 vaccine candidate currently under test. METHODOLOGY AND FINDINGS To address the HIV-1 variation, we designed a novel T cell immunogen, designated HIV(CONSV), by assembling the 14 most conserved regions of the HIV-1 proteome into one chimaeric protein. Each segment is a consensus sequence from one of the four major HIV-1 clades A, B, C and D, which alternate to ensure equal clade coverage. The gene coding for the HIV(CONSV) protein was inserted into the three most studied vaccine vectors, plasmid DNA, human adenovirus serotype 5 and modified vaccine virus Ankara (MVA), and induced HIV-1-specific T cell responses in mice. We also demonstrated that these conserved regions prime CD8(+) and CD4(+) T cell to highly conserved epitopes in humans and that these epitopes, although usually subdominant, generate memory T cells in patients during natural HIV-1 infection. SIGNIFICANCE Therefore, this vaccine approach provides an attractive and testable alternative for overcoming the HIV-1 variability, while focusing T cell responses on regions of the virus that are less likely to mutate and escape. Furthermore, this approach has merit in the simplicity of design and delivery, requiring only a single immunogen to provide extensive coverage of global HIV-1 population diversity.
Collapse
Affiliation(s)
- Sven Létourneau
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Eung-Jun Im
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Tumelo Mashishi
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Choechoe Brereton
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Anne Bridgeman
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Hongbing Yang
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Lucy Dorrell
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Tao Dong
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Bette Korber
- Los Alamo National Laboratory, Theoretical Biology and Biophysics, Los Alamos, New Mexico, United States of America
- The Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | - Andrew J. McMichael
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| | - Tomáš Hanke
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford, United Kingdom
| |
Collapse
|
94
|
Abstract
Synthetic peptide vaccines have potential to control viral infections. Successful experimental models using this approach include the protection of mice against the lethal Sendai virus infection by MHC class I binding CTL peptide epitope. The main benefit of vaccination with peptide epitopes is the ability to minimize the amount and complexity of a well-defined antigen. An appropriate peptide immunogen would also decrease the chance of stimulating a response against self-antigens, thereby providing a safer vaccine by avoiding autoimmunity. In general, the peptide vaccine strategy needs to dissect the specificity of antigen processing, the presence of B-and T-cell epitopes and the MHC restriction of the T-cell responses. This article briefly reviews the implications in the design of peptide vaccines and discusses the various approaches that are applied to improve their immunogenicity.
Collapse
Affiliation(s)
- Ali Azizi
- Variation Biotechnologies Inc., 22 de Varennes, Suite 210, Gatineau, QC J8T 8R1, Canada
| | | |
Collapse
|
95
|
Pallikkuth S, Wanchu A, Bhatnagar A, Sachdeva RK, Sharma M. Human immunodeficiency virus (HIV) gag antigen-specific T-helper and granule-dependent CD8 T-cell activities in exposed but uninfected heterosexual partners of HIV type 1-infected individuals in North India. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 14:1196-202. [PMID: 17823271 PMCID: PMC2043305 DOI: 10.1128/cvi.00488-06] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 02/23/2007] [Accepted: 06/21/2007] [Indexed: 11/20/2022]
Abstract
Repeated exposure to human immunodeficiency virus (HIV) does not always result in HIV infection, and several cohorts of HIV-exposed but uninfected (EU) individuals have been described. We studied T-helper and granule-dependent cytotoxic T-lymphocyte (CTL) activities in a group of 30 EU partners of HIV type 1 (HIV-1)-infected individuals. HIV-1-specific helper-T-cell activity was studied by measuring the levels of interleukin 2 (IL-2) produced by peripheral blood mononuclear cells (PBMCs) and the granule-dependent CTL activity by measuring the intracellular levels of perforin and granzyme B expression in CD8+ T cells after stimulation with gag p24 antigen. Elevated IL-2 production by PBMCs after p24 stimulation occurred in EU individuals. The levels of perforin and granzyme B expression in CD8+ T cells were also higher among EU individuals than among healthy controls. HIV-specific helper-T-cell and granule-dependent CTL activities inversely correlated with the time since the last unprotected sexual exposure in these individuals. In our cohort, activation of T-helper and granule-dependent CTL activities against HIV might be due to unprotected sexual contact. These results indicate that HIV-1-specific T-cell responses could play a role in protection against acquiring infection in this cohort of EU individuals.
Collapse
Affiliation(s)
- Suresh Pallikkuth
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | | | | | | | | |
Collapse
|
96
|
Tsegaye A, Ran L, Wolday D, Petros B, Dorigo W, Piriou E, Messele T, Sanders E, Tilahun T, Eshetu D, Schuitemaker H, Coutinho RA, Miedema F, Borghans J, van Baarle D. HIV-1 Subtype C gag-specific T-cell responses in relation to human leukocyte antigens in a diverse population of HIV-infected Ethiopians. J Acquir Immune Defic Syndr 2007; 45:389-400. [PMID: 17417101 DOI: 10.1097/qai.0b013e318059beaa] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Knowledge of the most dominant T-cell epitopes in the context of the local human leukocyte antigen (HLA) background is a prerequisite for the development of an effective HIV vaccine. In 100 Ethiopian subjects, 16 different HLA-A, 23 HLA-B, and 12 HLA-C specificities were observed. Ninety-four percent of the population carried at least 1 of the 5 most common HLA-A and/or HLA-B specificities. HIV-specific T-cell responses were measured in 48 HIV-infected Ethiopian subjects representing a wide range of ethnicities in Ethiopia using the interferon (IFN)-gamma enzyme-linked immunospot (Elispot) assay and 49 clade C-specific synthetic Gag peptides. Fifty-eight percent of the HIV-positive study subjects showed T-cell responses directed to 1 or more HIV Gag peptides. Most Gag-specific responses were directed against the subset of peptides spanning Gag p24. The breadth of response ranged from 1 to 9 peptides, with most (78%) individuals showing detectable responses to <3 Gag peptides. The magnitude of HIV-specific T-cell responses was not associated with HIV viral load but correlated positively with CD4 T-cell counts. The most frequently targeted Gag peptides overlapped with those previously described for HIV-1 subtype C-infected southern Africans, and therefore can be used in a multiethnic vaccine.
Collapse
Affiliation(s)
- Aster Tsegaye
- Ethiopian-Netherlands AIDS Research Project, Ethiopian Health and Nutrition Research Institute, Addis Ababa, Ethiopia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Patel V, Goodman A. Researching protective and promotive factors in mental health. Int J Epidemiol 2007; 36:703-7. [PMID: 17646185 DOI: 10.1093/ije/dym147] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
98
|
Abstract
The development of HIV-1 vaccines and microbicides remains hindered by our limited understanding of correlates of immune protection to infection. Evidence indicating that resistance to HIV-1 infection is indeed possible comes from HIV-1-exposed yet uninfected individuals, including cohorts of commercial sex workers and discordant couples. Despite their uninfected status some of these individuals have mucosal and systemic HIV-1-specific humoral and cellular immune responses in addition to their innate immune response. The combined contribution of innate and adaptive immunity as well as genetic factors is most likely of great importance for this protection against infection. Here we review data on the antibody responses and secreted immune molecules of the innate immune system in the female genital tract with emphasis on individuals who seem to resist HIV-1-infection despite repeated exposure to the virus.
Collapse
Affiliation(s)
- T Hirbod
- Center for Molecular Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
99
|
|
100
|
Schaubert KL, Price DA, Frahm N, Li J, Ng HL, Joseph A, Paul E, Majumder B, Ayyavoo V, Gostick E, Adams S, Marincola FM, Sewell AK, Altfeld M, Brenchley JM, Douek DC, Yang OO, Brander C, Goldstein H, Kan-Mitchell J. Availability of a diversely avid CD8+ T cell repertoire specific for the subdominant HLA-A2-restricted HIV-1 Gag p2419-27 epitope. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:7756-66. [PMID: 17548613 PMCID: PMC2365726 DOI: 10.4049/jimmunol.178.12.7756] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HLA-A2-restricted CTL responses to immunodominant HIV-1 epitopes do not appear to be very effective in the control of viral replication in vivo. In this study, we studied human CD8+ T cell responses to the subdominant HLA-A2-restricted epitope TV9 (Gag p24(19-27), TLNAWVKVV) to explore the possibility of increasing its immune recognition. We confirmed in a cohort of 313 patients, infected by clade B or clade C viruses, that TV9 is rarely recognized. Of interest, the functional sensitivity of the TV9 response can be relatively high. The potential T cell repertoires for TV9 and the characteristics of constituent clonotypes were assessed by ex vivo priming of circulating CD8+ T cells from healthy seronegative donors. TV9-specific CTLs capable of suppressing viral replication in vitro were readily generated, suggesting that the cognate T cell repertoire is not limiting. However, these cultures contained multiple discrete populations with a range of binding avidities for the TV9 tetramer and correspondingly distinct functional dependencies on the CD8 coreceptor. The lack of dominant clonotypes was not affected by the stage of maturation of the priming dendritic cells. Cultures primed by dendritic cells transduced to present endogenous TV9 were also incapable of clonal maturation. Thus, a diffuse TCR repertoire appeared to be an intrinsic characteristic of TV9-specific responses. These data indicate that subdominance is not a function of poor immunogenicity, cognate TCR repertoire availability, or the potential avidity properties thereof, but rather suggest that useful responses to this epitope are suppressed by competing CD8+ T cell populations during HIV-1 infection.
Collapse
Affiliation(s)
- Keri L. Schaubert
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Biological Sciences, University of Texas, El Paso, TX 79968
| | - David A. Price
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- Weatherall Institute of Molecular Medicine, University of Oxford, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Nicole Frahm
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Charles-town, MA 02192
| | - Jinzhu Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Hwee L. Ng
- Department of Medicine and AIDS Institute, Center for Health Sciences, University of California, Los Angeles, CA 90095
| | - Aviva Joseph
- Department of Micro-biology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Elyse Paul
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Biswanath Majumder
- Department of Infectious Diseases and Micro-biology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261
| | - Velpandi Ayyavoo
- Department of Infectious Diseases and Micro-biology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261
| | - Emma Gostick
- Weatherall Institute of Molecular Medicine, University of Oxford, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Sharon Adams
- Section of Immunogenetics, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD 20892
| | - Francesco M. Marincola
- Section of Immunogenetics, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD 20892
| | - Andrew K. Sewell
- Department of Medical Biochemistry and Immunology, Cardiff University, Cardiff, Wales, United Kingdom
| | - Marcus Altfeld
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Charles-town, MA 02192
| | - Jason M. Brenchley
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Otto O. Yang
- Department of Medicine and AIDS Institute, Center for Health Sciences, University of California, Los Angeles, CA 90095
| | - Christian Brander
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, Charles-town, MA 02192
| | - Harris Goldstein
- Department of Micro-biology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - June Kan-Mitchell
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Biological Sciences, University of Texas, El Paso, TX 79968
| |
Collapse
|