301
|
de Goede AL, Vulto AG, Osterhaus ADME, Gruters RA. Understanding HIV infection for the design of a therapeutic vaccine. Part II: Vaccination strategies for HIV. ANNALES PHARMACEUTIQUES FRANÇAISES 2014; 73:169-79. [PMID: 25528627 DOI: 10.1016/j.pharma.2014.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023]
Abstract
HIV infection leads to a gradual loss CD4(+) T lymphocytes comprising immune competence and progression to AIDS. Effective treatment with combined antiretroviral drugs (cART) decreases viral load below detectable levels but is not able to eliminate the virus from the body. The success of cART is frustrated by the requirement of expensive lifelong adherence, accumulating drug toxicities and chronic immune activation resulting in increased risk of several non-AIDS disorders, even when viral replication is suppressed. Therefore, there is a strong need for therapeutic strategies as an alternative to cART. Immunotherapy, or therapeutic vaccination, aims to increase existing immune responses against HIV or induce de novo immune responses. These immune responses should provide a functional cure by controlling viral replication and preventing disease progression in the absence of cART. The key difficulty in the development of an HIV vaccine is our ignorance of the immune responses that control of viral replication, and thus how these responses can be elicited and how they can be monitored. Part one of this review provides an extensive overview of the (patho-) physiology of HIV infection. It describes the structure and replication cycle of HIV, the epidemiology and pathogenesis of HIV infection and the innate and adaptive immune responses against HIV. Part two of this review discusses therapeutic options for HIV. Prevention modalities and antiretroviral therapy are briefly touched upon, after which an extensive overview on vaccination strategies for HIV is provided, including the choice of immunogens and delivery strategies.
Collapse
Affiliation(s)
- A L de Goede
- Department of Viroscience, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands; Department of Hospital Pharmacy, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands.
| | - A G Vulto
- Department of Hospital Pharmacy, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands
| | - A D M E Osterhaus
- Department of Viroscience, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands
| | - R A Gruters
- Department of Viroscience, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
302
|
Gordon SN, Doster MN, Kines RC, Keele BF, Brocca-Cofano E, Guan Y, Pegu P, Liyanage NPM, Vaccari M, Cuburu N, Buck CB, Ferrari G, Montefiori D, Piatak M, Lifson JD, Xenophontos AM, Venzon D, Robert-Guroff M, Graham BS, Lowy DR, Schiller JT, Franchini G. Antibody to the gp120 V1/V2 loops and CD4+ and CD8+ T cell responses in protection from SIVmac251 vaginal acquisition and persistent viremia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:6172-83. [PMID: 25398324 PMCID: PMC4335709 DOI: 10.4049/jimmunol.1401504] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The human papillomavirus pseudovirions (HPV-PsVs) approach is an effective gene-delivery system that can prime or boost an immune response in the vaginal tract of nonhuman primates and mice. Intravaginal vaccination with HPV-PsVs expressing SIV genes, combined with an i.m. gp120 protein injection, induced humoral and cellular SIV-specific responses in macaques. Priming systemic immune responses with i.m. immunization with ALVAC-SIV vaccines, followed by intravaginal HPV-PsV-SIV/gp120 boosting, expanded and/or recruited T cells in the female genital tract. Using a stringent repeated low-dose intravaginal challenge with the highly pathogenic SIVmac251, we show that although these regimens did not demonstrate significant protection from virus acquisition, they provided control of viremia in a number of animals. High-avidity Ab responses to the envelope gp120 V1/V2 region correlated with delayed SIVmac251 acquisition, whereas virus levels in mucosal tissues were inversely correlated with antienvelope CD4(+) T cell responses. CD8(+) T cell depletion in animals with controlled viremia caused an increase in tissue virus load in some animals, suggesting a role for CD8(+) T cells in virus control. This study highlights the importance of CD8(+) cells and antienvelope CD4(+) T cells in curtailing virus replication and antienvelope V1/V2 Abs in preventing SIVmac251 acquisition.
Collapse
Affiliation(s)
- Shari N Gordon
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD 20892
| | - Melvin N Doster
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD 20892
| | - Rhonda C Kines
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20982
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | | | - Yongjun Guan
- Division of Basic Science and Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Poonam Pegu
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD 20892
| | - Namal P M Liyanage
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD 20892
| | - Monica Vaccari
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD 20892
| | - Nicolas Cuburu
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20982
| | - Christopher B Buck
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20982
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, NC 27710
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Anastasia M Xenophontos
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD 20892
| | - David Venzon
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, MD 20892; and
| | | | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892
| | - Douglas R Lowy
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20982
| | - John T Schiller
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20982
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, Bethesda, MD 20892;
| |
Collapse
|
303
|
Strauss-Albee DM, Horowitz A, Parham P, Blish CA. Coordinated regulation of NK receptor expression in the maturing human immune system. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:4871-9. [PMID: 25288567 PMCID: PMC4225175 DOI: 10.4049/jimmunol.1401821] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
NK cells are responsible for recognizing and killing transformed, stressed, and infected cells. They recognize a set of non-Ag-specific features termed "altered self" through combinatorial signals from activating and inhibitory receptors. These NKRs are also expressed on CD4(+) and CD8(+) T cells, B cells, and monocytes, although a comprehensive inventory of NKR expression patterns across leukocyte lineages has never been performed. Using mass cytometry, we found that NKR expression patterns distinguish cell lineages in human peripheral blood. In individuals with high levels of CD57, indicative of a mature immune repertoire, NKRs are more likely to be expressed on non-NK cells, especially CD8(+) T cells. Mature NK and CD8(+) T cell populations show increased diversity of NKR surface expression patterns, but with distinct determinants: mature NK cells acquire primarily inhibitory receptors, whereas CD8(+) T cells attain a specific subset of both activating and inhibitory receptors, potentially imbuing them with a distinct functional role. Concurrently, monocytes show decreased expression of the generalized inhibitory receptor leukocyte Ig-like receptor subfamily b member 1, consistent with an increased activation threshold. Therefore, NKR expression is coordinately regulated as the immune system matures, resulting in the transfer of "altered self" recognition potential among leukocyte lineages. This likely reduces Ag specificity in the mature human immune system, and implies that vaccines and therapeutics that engage both its innate and adaptive branches may be more effective in the settings of aging and chronic infection.
Collapse
Affiliation(s)
- Dara M Strauss-Albee
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA 94305; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Amir Horowitz
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA 94305; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Peter Parham
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA 94305; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Catherine A Blish
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA 94305; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305;
| |
Collapse
|
304
|
Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM. Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat Rev Microbiol 2014; 12:750-64. [PMID: 25402363 PMCID: PMC4383747 DOI: 10.1038/nrmicro3352] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Effective antiretroviral therapy (ART) blunts viraemia, which enables HIV-1-infected individuals to control infection and live long, productive lives. However, HIV-1 infection remains incurable owing to the persistence of a viral reservoir that harbours integrated provirus within host cellular DNA. This latent infection is unaffected by ART and hidden from the immune system. Recent studies have focused on the development of therapies to disrupt latency. These efforts unmasked residual viral genomes and highlighted the need to enable the clearance of latently infected cells, perhaps via old and new strategies that improve the HIV-1-specific immune response. In this Review, we explore new approaches to eradicate established HIV-1 infection and avoid the burden of lifelong ART.
Collapse
Affiliation(s)
- Nancie M Archin
- Department of Medicine, University of North Carolina at Chapel Hill
| | - Julia Marsh Sung
- Department of Medicine, University of North Carolina at Chapel Hill
| | - Carolina Garrido
- Department of Medicine, University of North Carolina at Chapel Hill
| | | | - David M Margolis
- 1] Department of Medicine, University of North Carolina at Chapel Hill. [2] Department of Microbiology and Immunology, University of North Carolina at Chapel Hill. [3] Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
305
|
Abstract
This special issue is focused on DNA vaccines, marking the two decades since the first demonstration of pre-clinical protection was published in Science (Ulmer et al.; Heterologous protection against influenza by injection of DNA encoding a viral protein. 1993). This introductory article provides an overview of the field and highlights the observations of the articles in this special issue while placing them in the context of other recent publications.
Collapse
|
306
|
Tongo M, Burgers WA. Challenges in the design of a T cell vaccine in the context of HIV-1 diversity. Viruses 2014; 6:3968-90. [PMID: 25341662 PMCID: PMC4213573 DOI: 10.3390/v6103968] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/15/2014] [Accepted: 10/18/2014] [Indexed: 12/27/2022] Open
Abstract
The extraordinary variability of HIV-1 poses a major obstacle to vaccine development. The effectiveness of a vaccine is likely to vary dramatically in different populations infected with different HIV-1 subtypes, unless innovative vaccine immunogens are developed to protect against the range of HIV-1 diversity. Immunogen design for stimulating neutralizing antibody responses focuses on “breadth” – the targeting of a handful of highly conserved neutralizing determinants on the HIV-1 Envelope protein that can recognize the majority of viruses across all HIV-1 subtypes. An effective vaccine will likely require the generation of both broadly cross-neutralizing antibodies and non-neutralizing antibodies, as well as broadly cross-reactive T cells. Several approaches have been taken to design such broadly-reactive and cross-protective T cell immunogens. Artificial sequences have been designed that reduce the genetic distance between a vaccine strain and contemporary circulating viruses; “mosaic” immunogens extend this concept to contain multiple potential T cell epitope (PTE) variants; and further efforts attempt to focus T cell immunity on highly conserved regions of the HIV-1 genome. Thus far, a number of pre-clinical and early clinical studies have been performed assessing these new immunogens. In this review, the potential use of these new immunogens is explored.
Collapse
Affiliation(s)
- Marcel Tongo
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Virology, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, Division of Medical Virology, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| |
Collapse
|
307
|
John M, Gaudieri S. Influence of HIV and HCV on T cell antigen presentation and challenges in the development of vaccines. Front Microbiol 2014; 5:514. [PMID: 25352836 PMCID: PMC4195390 DOI: 10.3389/fmicb.2014.00514] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/12/2014] [Indexed: 12/11/2022] Open
Abstract
Some of the central challenges for developing effective vaccines against HIV and hepatitis C virus (HCV) are similar. Both infections are caused by small, highly mutable, rapidly replicating RNA viruses with the ability to establish long-term chronic pathogenic infection in human hosts. HIV has caused 60 million infections globally and HCV 180 million and both viruses may co-exist among certain populations by virtue of common blood-borne, sexual, or vertical transmission. Persistence of both pathogens is achieved by evasion of intrinsic, innate, and adaptive immune defenses but with some distinct mechanisms reflecting their differences in evolutionary history, replication characteristics, cell tropism, and visibility to mucosal versus systemic and hepatic immune responses. A potent and durable antibody and T cell response is a likely requirement of future HIV and HCV vaccines. Perhaps the single biggest difference between the two vaccine design challenges is that in HCV, a natural model of protective immunity can be found in those who resolve acute infection spontaneously. Such spontaneous resolvers exhibit durable and functional CD4+ and CD8+ T cell responses (Diepolder et al., 1995; Cooper et al., 1999; Thimme et al., 2001; Grakoui et al., 2003; Lauer et al., 2004; Schulze Zur Wiesch et al., 2012). However, frequent re-infection suggests partial or lack of protective immunity against heterologous HCV strains, possibly indicative of the degree of genetic diversity of circulating HCV genotypes and subtypes. There is no natural model of protective immunity in HIV, however, studies of “elite controllers,” or individuals who have durably suppressed levels of plasma HIV RNA without antiretroviral therapy, has provided the strongest evidence for CD8+ T cell responses in controlling viremia and limiting reservoir burden in established infection. Here we compare and contrast the specific mechanisms of immune evasion used by HIV and HCV, which subvert adaptive human leukocyte antigen (HLA)-restricted T cell immunity in natural infection, and the challenges these pose for designing effective preventative or therapeutic vaccines.
Collapse
Affiliation(s)
- Mina John
- Institute for Immunology and Infectious Diseases, Murdoch University Murdoch, WA, Australia ; Department of Clinical Immunology, PathWest Laboratory Medicine WA, Royal Perth Hospital Perth, WA, Australia
| | - Silvana Gaudieri
- Institute for Immunology and Infectious Diseases, Murdoch University Murdoch, WA, Australia ; School of Anatomy, Physiology and Human Biology, University of Western Australia Crawley, WA, Australia
| |
Collapse
|
308
|
Abstract
The ultimate solution to the global HIV-1 epidemic will probably require the development of a safe and effective vaccine. Multiple vaccine platforms have been evaluated in preclinical and clinical trials, but given the disappointing results of clinical efficacy studies so far, novel vaccine approaches are needed. In this Opinion article, we discuss the scientific basis and clinical potential of novel adenovirus and cytomegalovirus vaccine vectors for HIV-1 as two contrasting but potentially complementary vector approaches. Both of these vector platforms have demonstrated partial protection against stringent simian immunodeficiency virus challenges in rhesus monkeys using different immunological mechanisms.
Collapse
|
309
|
Abstract
Although some success was achieved in recent years in HIV prevention, an effective vaccine remains the means with the most potential of curtailing HIV-1 infections worldwide. Despite multiple failed attempts, a recent HIV vaccine regimen demonstrated modest protection from infection. Although the protective efficacy in this trial was not sufficient to warrant licensure, it spurred renewed optimism in the field and has provided valuable insights for improving future vaccine designs. This review summarizes the pertinent details of vaccine development and discusses ways the field is moving forward to develop a vaccine to prevent HIV infection and disease progression.
Collapse
Affiliation(s)
- Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 908, 20th Street South, CCB 328, Birmingham, AL 35294, USA.
| | - Anju Bansal
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 845, 19th Street South, BBRB 557, Birmingham, AL 35294, USA
| |
Collapse
|
310
|
Ensoli B, Cafaro A, Monini P, Marcotullio S, Ensoli F. Challenges in HIV Vaccine Research for Treatment and Prevention. Front Immunol 2014; 5:417. [PMID: 25250026 PMCID: PMC4157563 DOI: 10.3389/fimmu.2014.00417] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 08/18/2014] [Indexed: 12/17/2022] Open
Abstract
Many attempts have been made or are ongoing for HIV prevention and HIV cure. Many successes are in the list, particularly for HIV drugs, recently proposed also for prevention. However, no eradication of infection has been achieved so far with any drug. Further, a residual immune dysregulation associated to chronic immune activation and incomplete restoration of B and T cell subsets, together with HIV DNA persistence in reservoirs, are still unmet needs of the highly active antiretroviral therapy, causing novel “non-AIDS related” diseases that account for a higher risk of death even in virologically suppressed patients. These “ART unmet needs” represent a problem, which is expected to increase by ART roll out. Further, in countries such as South Africa, where six millions of individuals are infected, ART appears unable to contain the epidemics. Regretfully, all the attempts at developing a preventative vaccine have been largely disappointing. However, recent therapeutic immunization strategies have opened new avenues for HIV treatment, which might be exploitable also for preventative vaccine approaches. For example, immunization strategies aimed at targeting key viral products responsible of virus transmission, activation, and maintenance of virus reservoirs may intensify drug efficacy and lead to a functional cure providing new perspectives also for prevention and future virus eradication strategies. However, this approach imposes new challenges to the scientific community, vaccine developers, and regulatory bodies, such as the identification of novel immunological and virological biomarkers to assess efficacy end-points, taking advantage from the natural history of infection and exploiting lessons from former trials. This review will focus first on recent advancement of therapeutic strategies, then on the progresses made in preventative approaches, discussing concepts, and problems for the way ahead for the development of vaccines for HIV treatment and prevention.
Collapse
Affiliation(s)
- Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità , Rome , Italy
| | - Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità , Rome , Italy
| | - Paolo Monini
- National AIDS Center, Istituto Superiore di Sanità , Rome , Italy
| | | | - Fabrizio Ensoli
- Pathology and Microbiology, San Gallicano Institute, "Istituti Fisioterapici Ospitalieri" , Rome , Italy
| |
Collapse
|
311
|
Ishii D, Rosenblum JM, Nozaki T, Schenk AD, Setoguchi K, Su CA, Gorbacheva V, Baldwin WM, Valujskikh A, Fairchild RL. Novel CD8 T cell alloreactivities in CCR5-deficient recipients of class II MHC disparate kidney grafts. THE JOURNAL OF IMMUNOLOGY 2014; 193:3816-24. [PMID: 25172484 DOI: 10.4049/jimmunol.1303256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recipient CD4 T regulatory cells inhibit the acute T cell-mediated rejection of renal allografts in wild-type mice. The survival of single class II MHC-disparate H-2(bm12) renal allografts was tested in B6.CCR5(-/-) recipients, which have defects in T regulatory cell activities that constrain alloimmune responses. In contrast to wild-type C57BL/6 recipients, B6.CCR5(-/-) recipients rejected the bm12 renal allografts. However, donor-reactive CD8 T cells rather than CD4 T cells were the primary effector T cells mediating rejection. The CD8 T cells induced to bm12 allografts in CCR5-deficient recipients were reactive to peptides spanning the 3 aa difference in the I-A(bm12) versus I-A(b) β-chains presented by K(b) and D(b) class I MHC molecules. Allograft-primed CD8 T cells from CCR5-deficient allograft recipients were activated during culture either with proinflammatory cytokine-stimulated wild-type endothelial cells pulsed with the I-A(bm12) peptides or with proinflammatory cytokine-simulated bm12 endothelial cells, indicating their presentation of the I-A(bm12) β-chain peptide/class I MHC complexes. In addition to induction by bm12 renal allografts, the I-A(bm12) β-chain-reactive CD8 T cells were induced in CCR5-deficient, but not wild-type C57BL/6, mice by immunization with the peptides. These results reveal novel alloreactive CD8 T cell specificities in CCR5-deficient recipients of single class II MHC renal allografts that mediate rejection of the allografts.
Collapse
Affiliation(s)
- Daisuke Ishii
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Urology, Kitasato University, Kanagawa 228-8555, Japan
| | - Joshua M Rosenblum
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | - Taiji Nozaki
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Austin D Schenk
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | - Kiyoshi Setoguchi
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Charles A Su
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | | | - William M Baldwin
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and Glickman Urological Institute and the Transplant Center, Cleveland Clinic, Cleveland, OH 44195
| | - Anna Valujskikh
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and Glickman Urological Institute and the Transplant Center, Cleveland Clinic, Cleveland, OH 44195
| | - Robert L Fairchild
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and Glickman Urological Institute and the Transplant Center, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
312
|
Mechanisms of HIV protein degradation into epitopes: implications for vaccine design. Viruses 2014; 6:3271-92. [PMID: 25196483 PMCID: PMC4147695 DOI: 10.3390/v6083271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/06/2014] [Accepted: 08/11/2014] [Indexed: 12/02/2022] Open
Abstract
The degradation of HIV-derived proteins into epitopes displayed by MHC-I or MHC-II are the first events leading to the priming of HIV-specific immune responses and to the recognition of infected cells. Despite a wealth of information about peptidases involved in protein degradation, our knowledge of epitope presentation during HIV infection remains limited. Here we review current data on HIV protein degradation linking epitope production and immunodominance, viral evolution and impaired epitope presentation. We propose that an in-depth understanding of HIV antigen processing and presentation in relevant primary cells could be exploited to identify signatures leading to efficient or inefficient epitope presentation in HIV proteomes, and to improve the design of immunogens eliciting immune responses efficiently recognizing all infected cells.
Collapse
|
313
|
Brizić I, Lenac Roviš T, Krmpotić A, Jonjić S. MCMV avoidance of recognition and control by NK cells. Semin Immunopathol 2014; 36:641-50. [DOI: 10.1007/s00281-014-0441-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/15/2014] [Indexed: 01/27/2023]
|
314
|
Karl JA, Heimbruch KE, Vriezen CE, Mironczuk CJ, Dudley DM, Wiseman RW, O'Connor DH. Survey of major histocompatibility complex class II diversity in pig-tailed macaques. Immunogenetics 2014; 66:613-23. [PMID: 25129472 DOI: 10.1007/s00251-014-0797-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/11/2014] [Indexed: 12/21/2022]
Abstract
Pig-tailed macaques (Macaca nemestrina) serve as important models for human infectious disease research. Major histocompatibility complex (MHC) class II molecules are important to this research since they present peptides to CD4+ T cells. Despite the importance of characterizing the MHC-II alleles expressed in model species like pig-tailed macaques, to date, less than 150 MHC-II alleles have been named for the six most common classical class II loci (DRA, DRB, DQA, DQB, DPA, and DPB) in this population. Additionally, only a small percentage of these alleles are full-length, making it impossible to use the known sequence for reagent development. To address this, we developed a fast, high-throughput method to discover full-length MHC-II alleles and used it to characterize alleles in 32 pig-tailed macaques. By this method, we identified 128 total alleles across all six loci. We also performed an exon 2-based genotyping assay to validate the full-length sequencing results; this genotyping assay could be optimized for use in determining MHC-II allele frequencies in large cohorts of pig-tailed macaques.
Collapse
Affiliation(s)
- Julie A Karl
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | | | | | | | | | | | | |
Collapse
|
315
|
Tuero I, Robert-Guroff M. Challenges in mucosal HIV vaccine development: lessons from non-human primate models. Viruses 2014; 6:3129-58. [PMID: 25196380 PMCID: PMC4147690 DOI: 10.3390/v6083129] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 12/23/2022] Open
Abstract
An efficacious HIV vaccine is urgently needed to curb the AIDS pandemic. The modest protection elicited in the phase III clinical vaccine trial in Thailand provided hope that this goal might be achieved. However, new approaches are necessary for further advances. As HIV is transmitted primarily across mucosal surfaces, development of immunity at these sites is critical, but few clinical vaccine trials have targeted these sites or assessed vaccine-elicited mucosal immune responses. Pre-clinical studies in non-human primate models have facilitated progress in mucosal vaccine development by evaluating candidate vaccine approaches, developing methodologies for collecting and assessing mucosal samples, and providing clues to immune correlates of protective immunity for further investigation. In this review we have focused on non-human primate studies which have provided important information for future design of vaccine strategies, targeting of mucosal inductive sites, and assessment of mucosal immunity. Knowledge gained in these studies will inform mucosal vaccine design and evaluation in human clinical trials.
Collapse
Affiliation(s)
- Iskra Tuero
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Marjorie Robert-Guroff
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
316
|
Developments in Viral Vector-Based Vaccines. Vaccines (Basel) 2014; 2:624-41. [PMID: 26344749 PMCID: PMC4494222 DOI: 10.3390/vaccines2030624] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/18/2014] [Accepted: 06/30/2014] [Indexed: 12/22/2022] Open
Abstract
Viral vectors are promising tools for gene therapy and vaccines. Viral vector-based vaccines can enhance immunogenicity without an adjuvant and induce a robust cytotoxic T lymphocyte (CTL) response to eliminate virus-infected cells. During the last several decades, many types of viruses have been developed as vaccine vectors. Each has unique features and parental virus-related risks. In addition, genetically altered vectors have been developed to improve efficacy and safety, reduce administration dose, and enable large-scale manufacturing. To date, both successful and unsuccessful results have been reported in clinical trials. These trials provide important information on factors such as toxicity, administration dose tolerated, and optimized vaccination strategy. This review highlights major viral vectors that are the best candidates for clinical use.
Collapse
|
317
|
Alexandre YO, Cocita CD, Ghilas S, Dalod M. Deciphering the role of DC subsets in MCMV infection to better understand immune protection against viral infections. Front Microbiol 2014; 5:378. [PMID: 25120535 PMCID: PMC4114203 DOI: 10.3389/fmicb.2014.00378] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022] Open
Abstract
Infection of mice with murine cytomegalovirus (MCMV) recapitulates many physiopathological characteristics of human CMV infection and enables studying the interactions between a virus and its natural host. Dendritic cells (DC) are mononuclear phagocytes linking innate and adaptive immunity which are both necessary for MCMV control. DC are critical for the induction of cellular immunity because they are uniquely efficient for the activation of naïve T cells during their first encounter with a pathogen. DC are equipped with a variety of innate immune recognition receptors (I2R2) allowing them to detect pathogens or infections and to engulf molecules, microorganisms or cellular debris. The combinatorial engagement of I2R2 during infections controls DC maturation and shapes their response in terms of cytokine production, activation of natural killer (NK) cells and functional polarization of T cells. Several DC subsets exist which express different arrays of I2R2 and are specialized in distinct functions. The study of MCMV infection helped deciphering the physiological roles of DC subsets and their molecular regulation. It allowed the identification and first in vivo studies of mouse plasmacytoid DC which produce high level of interferons-α/β early after infection. Despite its ability to infect DC and dampen their functions, MCMV induces very robust, efficient and long-lasting CD8 T cell responses. Their priming may rely on the unique ability of uninfected XCR1+ DC to cross-present engulfed viral antigens and thus to counter MCMV interference with antigen presentation. A balance appears to have been reached during co-evolution, allowing controlled replication of the virus for horizontal spread without pathological consequences for the immunocompetent host. We will discuss the role of the interplay between the virus and DC in setting this balance, and how advancing this knowledge further could help develop better vaccines against other intracellular infectious agents.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Clément D Cocita
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Sonia Ghilas
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| |
Collapse
|
318
|
Beverley PCL, Ruzsics Z, Hey A, Hutchings C, Boos S, Bolinger B, Marchi E, O'Hara G, Klenerman P, Koszinowski UH, Tchilian EZ. A novel murine cytomegalovirus vaccine vector protects against Mycobacterium tuberculosis. THE JOURNAL OF IMMUNOLOGY 2014; 193:2306-16. [PMID: 25070842 PMCID: PMC4134927 DOI: 10.4049/jimmunol.1302523] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tuberculosis remains a global health problem so that a more effective vaccine than bacillus Calmette–Guérin is urgently needed. Cytomegaloviruses persist lifelong in vivo and induce powerful immune and increasing (“inflationary”) responses, making them attractive vaccine vectors. We have used an m1–m16-deleted recombinant murine CMV (MCMV) expressing Mycobacterium tuberculosis Ag 85A to show that infection of mice with this recombinant significantly reduces the mycobacterial load after challenge with M. tuberculosis, whereas control empty virus has a lesser effect. Both viruses induce immune responses to H-2d–restricted epitopes of MCMV pp89 and M18 Ags characteristic of infection with other MCMVs. A low frequency of 85A-specific memory cells could be revealed by in vivo or in vitro boosting or after challenge with M. tuberculosis. Kinetic analysis of M. tuberculosis growth in the lungs of CMV-infected mice shows early inhibition of M. tuberculosis growth abolished by treatment with NK-depleting anti–asialo ganglio-N-tetraosylceramide Ab. Microarray analysis of the lungs of naive and CMV-infected mice shows increased IL-21 mRNA in infected mice, whereas in vitro NK assays indicate increased levels of NK activity. These data indicate that activation of NK cells by MCMV provides early nonspecific protection against M. tuberculosis, potentiated by a weak 85A-specific T cell response, and they reinforce the view that the innate immune system plays an important role in both natural and vaccine-induced protection against M. tuberculosis.
Collapse
Affiliation(s)
- Peter C L Beverley
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Zsolt Ruzsics
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Ariann Hey
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Claire Hutchings
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Simone Boos
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Beatrice Bolinger
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Emanuele Marchi
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Geraldine O'Hara
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Ulrich H Koszinowski
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Elma Z Tchilian
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| |
Collapse
|
319
|
Sicurella M, Nicoli F, Gallerani E, Volpi I, Berto E, Finessi V, Destro F, Manservigi R, Cafaro A, Ensoli B, Caputo A, Gavioli R, Marconi PC. An attenuated herpes simplex virus type 1 (HSV1) encoding the HIV-1 Tat protein protects mice from a deadly mucosal HSV1 challenge. PLoS One 2014; 9:e100844. [PMID: 25033084 PMCID: PMC4102458 DOI: 10.1371/journal.pone.0100844] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 05/30/2014] [Indexed: 12/22/2022] Open
Abstract
Herpes simplex virus types 1 and 2 (HSV1 and HSV2) are common infectious agents in both industrialized and developing countries. They cause recurrent asymptomatic and/or symptomatic infections, and life-threatening diseases and death in newborns and immunocompromised patients. Current treatment for HSV relies on antiviral medications, which can halt the symptomatic diseases but cannot prevent the shedding that occurs in asymptomatic patients or, consequently, the spread of the viruses. Therefore, prevention rather than treatment of HSV infections has long been an area of intense research, but thus far effective anti-HSV vaccines still remain elusive. One of the key hurdles to overcome in anti-HSV vaccine development is the identification and effective use of strategies that promote the emergence of Th1-type immune responses against a wide range of epitopes involved in the control of viral replication. Since the HIV1 Tat protein has several immunomodulatory activities and increases CTL recognition of dominant and subdominant epitopes of heterologous antigens, we generated and assayed a recombinant attenuated replication-competent HSV1 vector containing the tat gene (HSV1-Tat). In this proof-of-concept study we show that immunization with this vector conferred protection in 100% of mice challenged intravaginally with a lethal dose of wild-type HSV1. We demonstrate that the presence of Tat within the recombinant virus increased and broadened Th1-like and CTL responses against HSV-derived T-cell epitopes and elicited in most immunized mice detectable IgG responses. In sharp contrast, a similarly attenuated HSV1 recombinant vector without Tat (HSV1-LacZ), induced low and different T cell responses, no measurable antibody responses and did not protect mice against the wild-type HSV1 challenge. These findings strongly suggest that recombinant HSV1 vectors expressing Tat merit further investigation for their potential to prevent and/or contain HSV1 infection and dissemination.
Collapse
Affiliation(s)
- Mariaconcetta Sicurella
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Francesco Nicoli
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Eleonora Gallerani
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Ilaria Volpi
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| | - Elena Berto
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| | - Valentina Finessi
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Federica Destro
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Roberto Manservigi
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| | - Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Caputo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Riccardo Gavioli
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Peggy C. Marconi
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
320
|
TMEM129 is a Derlin-1 associated ERAD E3 ligase essential for virus-induced degradation of MHC-I. Proc Natl Acad Sci U S A 2014; 111:11425-30. [PMID: 25030448 DOI: 10.1073/pnas.1409099111] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The US11 gene product of human cytomegalovirus promotes viral immune evasion by hijacking the endoplasmic reticulum (ER)-associated degradation (ERAD) pathway. US11 initiates dislocation of newly translocated MHC I from the ER to the cytosol for proteasome-mediated degradation. Despite the critical role for ubiquitin in this degradation pathway, the responsible E3 ligase is unknown. In a forward genetic screen for host ERAD components hijacked by US11 in near-haploid KBM7 cells, we identified TMEM129, an uncharacterized polytopic membrane protein. TMEM129 is essential and rate-limiting for US11-mediated MHC-I degradation and acts as a novel ER resident E3 ubiquitin ligase. TMEM129 contains an unusual cysteine-only RING with intrinsic E3 ligase activity and is recruited to US11 via Derlin-1. Together with its E2 conjugase Ube2J2, TMEM129 is responsible for the ubiquitination, dislocation, and subsequent degradation of US11-associated MHC-I. US11 engages two degradation pathways: a Derlin-1/TMEM129-dependent pathway required for MHC-I degradation and a SEL1L/HRD1-dependent pathway required for "free" US11 degradation. Our data show that TMEM129 is a novel ERAD E3 ligase and the central component of a novel mammalian ERAD complex.
Collapse
|
321
|
Esparza J, Van Regenmortel MHV. More Surprises in the Development of an HIV Vaccine. Front Immunol 2014; 5:329. [PMID: 25071786 PMCID: PMC4095567 DOI: 10.3389/fimmu.2014.00329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 06/29/2014] [Indexed: 01/16/2023] Open
Affiliation(s)
- José Esparza
- Institute of Human Virology, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Marc H V Van Regenmortel
- CNRS, UMR7242-Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), Université de Strasbourg , Illkirch-Graffenstaden , France
| |
Collapse
|
322
|
Abstract
Antiretroviral therapy (ART) is able to suppress HIV-1 replication indefinitely in individuals who have access to these medications, are able to tolerate these drugs, and are motivated to take them daily for life. However, ART is not curative. HIV-1 persists indefinitely during ART as quiescent integrated DNA within memory CD4(+) T cells and perhaps other long-lived cellular reservoirs. In this Review, we discuss the role of the immune system in the establishment and maintenance of the latent HIV-1 reservoir. A detailed understanding of how the host immune system shapes the size and distribution of the viral reservoir should lead to the development of a new generation of immune-based therapeutics, which may eventually contribute to a curative intervention.
Collapse
Affiliation(s)
- Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Steven G Deeks
- University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
323
|
Picker LJ. Are effector memory T cells the key to an effective HIV/AIDS vaccine? EMBO Rep 2014; 15:820-1. [PMID: 24980866 DOI: 10.15252/embr.201439052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
324
|
Haynes BF, Moody MA, Alam M, Bonsignori M, Verkoczy L, Ferrari G, Gao F, Tomaras GD, Liao HX, Kelsoe G. Progress in HIV-1 vaccine development. J Allergy Clin Immunol 2014; 134:3-10; quiz 11. [PMID: 25117798 DOI: 10.1016/j.jaci.2014.04.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/11/2014] [Accepted: 04/11/2014] [Indexed: 11/27/2022]
Abstract
The past 2 years have seen a number of basic and translational science advances in the quest for development of an effective HIV-1 vaccine. These advances include discovery of new envelope targets of potentially protective antibodies, demonstration that CD8(+) T cells can control HIV-1 infection, development of immunogens to overcome HIV-1 T-cell epitope diversity, identification of correlates of transmission risk in an HIV-1 efficacy trial, and mapping of the coevolution of HIV-1 founder envelope mutants in infected subjects with broad neutralizing antibodies, thereby defining broad neutralizing antibody developmental pathways. Despite these advances, a promising HIV-1 vaccine efficacy trial published in 2013 did not prevent infection, and the HIV-1 vaccine field is still years away from deployment of an effective vaccine. This review summarizes what some of the scientific advances have been, what roadblocks still remain, and what the most promising approaches are for progress in design of successful HIV-1 vaccine candidates.
Collapse
Affiliation(s)
- Barton F Haynes
- Duke Human Vaccine Institute, Durham, NC; Department of Medicine, Duke University School of Medicine, Durham, NC; Department of Immunology, Duke University School of Medicine, Durham, NC.
| | - M Anthony Moody
- Duke Human Vaccine Institute, Durham, NC; Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Munir Alam
- Duke Human Vaccine Institute, Durham, NC; Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Mattia Bonsignori
- Duke Human Vaccine Institute, Durham, NC; Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Laurent Verkoczy
- Duke Human Vaccine Institute, Durham, NC; Department of Pathology, Duke University School of Medicine, Durham, NC
| | - Guido Ferrari
- Duke Human Vaccine Institute, Durham, NC; Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Feng Gao
- Duke Human Vaccine Institute, Durham, NC; Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Durham, NC; Department of Immunology, Duke University School of Medicine, Durham, NC; Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Durham, NC; Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Garnett Kelsoe
- Duke Human Vaccine Institute, Durham, NC; Department of Immunology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
325
|
Therapeutic HIV vaccines: prior setbacks, current advances, and future prospects. Vaccine 2014; 32:5540-5. [PMID: 24968157 DOI: 10.1016/j.vaccine.2014.06.066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/30/2014] [Accepted: 06/12/2014] [Indexed: 01/17/2023]
|
326
|
Vargas-Inchaustegui DA, Tuero I, Mohanram V, Musich T, Pegu P, Valentin A, Sui Y, Rosati M, Bear J, Venzon DJ, Kulkarni V, Alicea C, Pilkington GR, Liyanage NPM, Demberg T, Gordon SN, Wang Y, Hogg AE, Frey B, Patterson LJ, DiPasquale J, Montefiori DC, Sardesai NY, Reed SG, Berzofsky JA, Franchini G, Felber BK, Pavlakis GN, Robert-Guroff M. Humoral immunity induced by mucosal and/or systemic SIV-specific vaccine platforms suggests novel combinatorial approaches for enhancing responses. Clin Immunol 2014; 153:308-22. [PMID: 24907411 DOI: 10.1016/j.clim.2014.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/13/2014] [Accepted: 05/21/2014] [Indexed: 12/22/2022]
Abstract
Combinatorial HIV/SIV vaccine approaches targeting multiple arms of the immune system might improve protective efficacy. We compared SIV-specific humoral immunity induced in rhesus macaques by five vaccine regimens. Systemic regimens included ALVAC-SIVenv priming and Env boosting (ALVAC/Env); DNA immunization; and DNA plus Env co-immunization (DNA&Env). RepAd/Env combined mucosal replication-competent Ad-env priming with systemic Env boosting. A Peptide/Env regimen, given solely intrarectally, included HIV/SIV peptides followed by MVA-env and Env boosts. Serum antibodies mediating neutralizing, phagocytic and ADCC activities were induced by ALVAC/Env, RepAd/Env and DNA&Env vaccines. Memory B cells and plasma cells were maintained in the bone marrow. RepAd/Env vaccination induced early SIV-specific IgA in rectal secretions before Env boosting, although mucosal IgA and IgG responses were readily detected at necropsy in ALVAC/Env, RepAd/Env, DNA&Env and DNA vaccinated animals. Our results suggest that combined RepAd priming with ALVAC/Env or DNA&Env regimen boosting might induce potent, functional, long-lasting systemic and mucosal SIV-specific antibodies.
Collapse
Affiliation(s)
- Diego A Vargas-Inchaustegui
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Iskra Tuero
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Venkatramanan Mohanram
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Thomas Musich
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Poonam Pegu
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - Yongjun Sui
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - David J Venzon
- Biostatistics and Data Management Section, CCR, NCI, NIH, Rockville, MD 20850, United States
| | - Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - Guy R Pilkington
- Human Retrovirus Pathogenesis Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - Namal P M Liyanage
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Thorsten Demberg
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Shari N Gordon
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Yichuan Wang
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Alison E Hogg
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Blake Frey
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - L Jean Patterson
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Janet DiPasquale
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - David C Montefiori
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC 27710, United States
| | | | - Steven G Reed
- Infectious Diseases Research Institute, Seattle, WA 98102, United States
| | - Jay A Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccine Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, CCR, NCI, NIH, Frederick, MD 21702, United States
| | - Marjorie Robert-Guroff
- Immune Biology of Retroviral Infection Section, Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
327
|
Saez-Cirion A, Jacquelin B, Barré-Sinoussi F, Müller-Trutwin M. Immune responses during spontaneous control of HIV and AIDS: what is the hope for a cure? Philos Trans R Soc Lond B Biol Sci 2014; 369:20130436. [PMID: 24821922 PMCID: PMC4024229 DOI: 10.1098/rstb.2013.0436] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
HIV research has made rapid progress and led to remarkable achievements in recent decades, the most important of which are combination antiretroviral therapies (cART). However, in the absence of a vaccine, the pandemic continues, and additional strategies are needed. The 'towards an HIV cure' initiative aims to eradicate HIV or at least bring about a lasting remission of infection during which the host can control viral replication in the absence of cART. Cases of spontaneous and treatment-induced control of infection offer substantial hope. Here, we describe the scientific knowledge that is lacking, and the priorities that have been established for research into a cure. We discuss in detail the immunological lessons that can be learned by studying natural human and animal models of protection and spontaneous control of viraemia or of disease progression. In particular, we describe the insights we have gained into the immune mechanisms of virus control, the impact of early virus-host interactions and why chronic inflammation, a hallmark of HIV infection, is an obstacle to a cure. Finally, we enumerate current interventions aimed towards improving the host immune response.
Collapse
Affiliation(s)
| | | | | | - M. Müller-Trutwin
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France
| |
Collapse
|
328
|
Opening Fronts in HIV Vaccine Development: Targeting reservoirs to clear and cure. Nat Med 2014; 20:480-1. [DOI: 10.1038/nm.3550] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
329
|
Human cytomegalovirus and autoimmune disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:472978. [PMID: 24967373 PMCID: PMC4022258 DOI: 10.1155/2014/472978] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/17/2014] [Indexed: 11/28/2022]
Abstract
Human cytomegalovirus (HCMV) represents a prototypic pathogenic member of the β-subgroup of the herpesvirus family. A range of HCMV features like its lytic replication in multiple tissues, the lifelong persistence through periods of latency and intermitting reactivation, the extraordinary large proteome, and extensive manipulation of adaptive and innate immunity make HCMV a high profile candidate for involvement in autoimmune disorders. We surveyed the available literature for reports on HCMV association with onset or exacerbation of autoimmune disease. A causative linkage between HCMV and systemic lupus erythematosus (SLE), systemic sclerosis (SSc), diabetes mellitus type 1, and rheumatoid arthritis (RA) is suggested by the literature. However, a clear association of HCMV seroprevalence and disease could not be established, leaving the question open whether HCMV could play a coresponsible role for onset of disease. For convincing conclusions population-based prospective studies must be performed in the future. Specific immunopathogenic mechanisms by which HCMV could contribute to the course of autoimmune disease have been suggested, for example, molecular mimicry by UL94 in SSc and UL83/pp65 in SLE patients, as well as aggravation of joint inflammation by induction and expansion of CD4+/CD28− T-cells in RA patients. Further studies are needed to validate these findings and to lay the grounds for targeted therapeutic intervention.
Collapse
|
330
|
Sambor A, Garcia A, Berrong M, Pickeral J, Brown S, Rountree W, Sanchez A, Pollara J, Frahm N, Keinonen S, Kijak GH, Roederer M, Levine G, D'Souza MP, Jaimes M, Koup R, Denny T, Cox J, Ferrari G. Establishment and maintenance of a PBMC repository for functional cellular studies in support of clinical vaccine trials. J Immunol Methods 2014; 409:107-16. [PMID: 24787274 DOI: 10.1016/j.jim.2014.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/28/2014] [Accepted: 04/08/2014] [Indexed: 11/19/2022]
Abstract
A large repository of cryopreserved peripheral blood mononuclear cells (PBMCs) samples was created to provide laboratories testing the specimens from human immunodeficiency virus-1 (HIV-1) vaccine clinical trials the material for assay development, optimization, and validation. One hundred thirty-one PBMC samples were collected using leukapheresis procedure between 2007 and 2013 by the Comprehensive T cell Vaccine Immune Monitoring Consortium core repository. The donors included 83 human immunodeficiency virus-1 (HIV-1) seronegative and 32 HIV-1 seropositive subjects. The samples were extensively characterized for the ability of T cell subsets to respond to recall viral antigens including cytomegalovirus, Epstein-Barr virus, influenza virus, and HIV-1 using Interferon-gamma (IFN-γ) enzyme linked immunospot (ELISpot) and IFN-γ/interleukin 2 (IL-2) intracellular cytokine staining (ICS) assays. A subset of samples was evaluated over time to determine the integrity of the cryopreserved samples in relation to recovery, viability, and functionality. The principal results of our study demonstrate that viable and functional cells were consistently recovered from the cryopreserved samples. Therefore, we determined that this repository of large size cryopreserved cellular samples constitutes a unique resource for laboratories that are involved in optimization and validation of assays to evaluate T, B, and NK cellular functions in the context of clinical trials.
Collapse
Affiliation(s)
- Anna Sambor
- Foundation for National Institutes of Health, Bethesda, MD, USA
| | - Ambrosia Garcia
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | | | | | - Sara Brown
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Wes Rountree
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | - Ana Sanchez
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | | | - Nicole Frahm
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sarah Keinonen
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | - Gustavo H Kijak
- Viral Genetics Section, US Military HIV Research Program, Henry M Jackson Foundation for the Advancement of Military Medicine, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | - Gail Levine
- Foundation for National Institutes of Health, Bethesda, MD, USA
| | | | | | - Richard Koup
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Thomas Denny
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA
| | - Josephine Cox
- International AIDS Vaccine Initiative, New York, NY, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
331
|
|
332
|
Mocarski ES, Kaiser WJ, Livingston-Rosanoff D, Upton JW, Daley-Bauer LP. True grit: programmed necrosis in antiviral host defense, inflammation, and immunogenicity. THE JOURNAL OF IMMUNOLOGY 2014; 192:2019-26. [PMID: 24563506 DOI: 10.4049/jimmunol.1302426] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Programmed necrosis mediated by receptor interacting protein kinase (RIP)3 (also called RIPK3) has emerged as an alternate death pathway triggered by TNF family death receptors, pathogen sensors, IFNRs, Ag-specific TCR activation, and genotoxic stress. Necrosis leads to cell leakage and acts as a "trap door," eliminating cells that cannot die by apoptosis because of the elaboration of pathogen-encoded caspase inhibitors. Necrotic signaling requires RIP3 binding to one of three partners-RIP1, DAI, or TRIF-via a common RIP homotypic interaction motif. Once activated, RIP3 kinase targets the pseudokinase mixed lineage kinase domain-like to drive cell lysis. Although necrotic and apoptotic death can enhance T cell cross-priming during infection, mice that lack these extrinsic programmed cell death pathways are able to produce Ag-specific T cells and control viral infection. The entwined relationship of apoptosis and necrosis evolved in response to pathogen-encoded suppressors to support host defense and contribute to inflammation.
Collapse
Affiliation(s)
- Edward S Mocarski
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | | | | | | | | |
Collapse
|
333
|
Malouli D, Hansen SG, Nakayasu ES, Marshall EE, Hughes CM, Ventura AB, Gilbride RM, Lewis MS, Xu G, Kreklywich C, Whizin N, Fischer M, Legasse AW, Viswanathan K, Siess D, Camp DG, Axthelm MK, Kahl C, DeFilippis VR, Smith RD, Streblow DN, Picker LJ, Früh K. Cytomegalovirus pp65 limits dissemination but is dispensable for persistence. J Clin Invest 2014; 124:1928-44. [PMID: 24691437 DOI: 10.1172/jci67420] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 02/13/2014] [Indexed: 11/17/2022] Open
Abstract
The most abundantly produced virion protein in human cytomegalovirus (HCMV) is the immunodominant phosphoprotein 65 (pp65), which is frequently included in CMV vaccines. Although it is nonessential for in vitro CMV growth, pp65 displays immunomodulatory functions that support a potential role in primary and/or persistent infection. To determine the contribution of pp65 to CMV infection and immunity, we generated a rhesus CMV lacking both pp65 orthologs (RhCMVΔpp65ab). While deletion of pp65ab slightly reduced growth in vitro and increased defective particle formation, the protein composition of secreted virions was largely unchanged. Interestingly, pp65 was not required for primary and persistent infection in animals. Immune responses induced by RhCMVΔpp65ab did not prevent reinfection with rhesus CMV; however, reinfection with RhCMVΔUS2-11, which lacks viral-encoded MHC-I antigen presentation inhibitors, was prevented. Unexpectedly, induction of pp65b-specific T cells alone did not protect against RhCMVΔUS2-11 challenge, suggesting that T cells targeting multiple CMV antigens are required for protection. However, pp65-specific immunity was crucial for controlling viral dissemination during primary infection, as indicated by the marked increase of RhCMVΔpp65ab genome copies in CMV-naive, but not CMV-immune, animals. Our data provide rationale for inclusion of pp65 into CMV vaccines but also demonstrate that pp65-induced T cell responses alone do not recapitulate the protective effect of natural infection.
Collapse
|
334
|
McMichael AJ, Koff WC. Vaccines that stimulate T cell immunity to HIV-1: the next step. Nat Immunol 2014; 15:319-22. [PMID: 24646598 PMCID: PMC4324504 DOI: 10.1038/ni.2844] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/05/2014] [Indexed: 12/20/2022]
Abstract
The search for a vaccine against human immunodeficiency virus type 1 (HIV-1) has many hurdles to overcome. Ideally, the stimulation of both broadly neutralizing antibodies and cell-mediated immune responses remains the best option, but no candidate in clinical trials at present has elicited such antibodies, and efficacy trials have not demonstrated any benefit for vaccines designed to stimulate immune responses of CD8(+) T cells. Findings obtained with the simian immunodeficiency virus (SIV) monkey model have provided new evidence that stimulating effective CD8(+) T cell immunity could provide protection, and in this Perspective we explore the path forward for optimizing such responses in humans.
Collapse
Affiliation(s)
| | - Wayne C Koff
- International AIDS Vaccine Initiative, New York, New York, USA
| |
Collapse
|
335
|
McMichael A, Dorrell L. Comment on clinical development of candidate HIV vaccines: different problems for different vaccines. AIDS Res Hum Retroviruses 2014; 30:331-2. [PMID: 24606555 DOI: 10.1089/aid.2014.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Andrew McMichael
- 1 Weatherall Institute of Molecular Medicine, University of Oxford , Oxford, United Kingdom
| | | |
Collapse
|
336
|
Passaes CP, Sáez-Cirión A. HIV cure research: advances and prospects. Virology 2014; 454-455:340-52. [PMID: 24636252 DOI: 10.1016/j.virol.2014.02.021] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 12/16/2022]
Abstract
Thirty years after the identification of HIV, a cure for HIV infection is still to be achieved. Advances of combined antiretroviral therapy (cART) in recent years have transformed HIV infection into a chronic disease when treatment is available. However, in spite of the favorable outcomes provided by the newer therapies, cART is not curative and patients are at risk of developing HIV-associated disorders. Moreover, universal access to antiretroviral treatment is restricted by financial obstacles. This review discusses the most recent strategies that have been developed in the search for an HIV cure and to improve life quality of people living with HIV.
Collapse
Affiliation(s)
- Caroline P Passaes
- Unité de Régulation des Infections Rétrovirales, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France; CEA, Division of Immuno-Virology, iMETI/DSV, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Asier Sáez-Cirión
- Unité de Régulation des Infections Rétrovirales, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
337
|
Foley MH, Forcier T, McAndrew E, Gonzalez M, Chen H, Juelg B, Walker BD, Irvine DJ. High avidity CD8+ T cells efficiently eliminate motile HIV-infected targets and execute a locally focused program of anti-viral function. PLoS One 2014; 9:e87873. [PMID: 24551068 PMCID: PMC3923750 DOI: 10.1371/journal.pone.0087873] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/29/2013] [Indexed: 11/17/2022] Open
Abstract
The dissemination of HIV from an initial site of infection is facilitated by motile HIV-infected CD4+ T-cells. However, the impact of infected target cell migration on antigen recognition by HIV-specific CD8+ T-cells is unclear. Using a 3D in vitro model of tissue, we visualized dynamic interactions between HIV-infected or peptide-pulsed CD4+ T-cells and HIV-specific CD8+ T-cells. CTLs engaged motile HIV-infected targets, but ∼50% of targets broke contact and escaped. In contrast, immobilized target cells were readily killed, indicating target motility directly inhibits CD8+ T-cell function. Strong calcium signals occurred in CTLs killing a motile target but calcium signaling was weak or absent in CTLs which permitted target escape. Neutralization of adhesion receptors LFA-1 and CD58 inhibited CD8+ T-cell function within the 3D matrix, demonstrating that efficient motile target lysis as dependent on adhesive engagement of targets. Antigen sensitivity (a convolution of antigen density, TCR avidity and CD8 coreceptor binding) is also critical for target recognition. We modulated this parameter (known as functional avidity but referred to here as “avidity” for the sake of simplicity) by exploiting common HIV escape mutations and measured their impact on CTL function at the single-cell level. Targets pulsed with low avidity mutant antigens frequently escaped while CTLs killed targets bearing high avidity antigen with near-perfect efficiency. CTLs engaged, arrested, and killed an initial target bearing high avidity antigen within minutes, but serial killing was surprisingly rare. CD8 cells remained committed to their initial dead target for hours, accumulating TCR signals that sustained secretion of soluble antiviral factors. These data indicate that high-avidity CD8+ T-cells execute an antiviral program in the precise location where antigen has been sensed: CTL effector functions are spatiotemporally coordinated with an early lytic phase followed by a sustained stationary secretory phase to control local viral infection.
Collapse
Affiliation(s)
- Maria Hottelet Foley
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America ; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America ; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Talitha Forcier
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America ; Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Elizabeth McAndrew
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Michael Gonzalez
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Huabiao Chen
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Boris Juelg
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America
| | - Bruce D Walker
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America ; Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America ; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, Massachusetts, United States of America ; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America ; Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America ; Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| |
Collapse
|
338
|
Hanke T. Conserved immunogens in prime-boost strategies for the next-generation HIV-1 vaccines. Expert Opin Biol Ther 2014; 14:601-16. [PMID: 24490585 DOI: 10.1517/14712598.2014.885946] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Effective vaccines are the best solution for stopping the spread of HIV/AIDS and other infectious diseases. Their development and in-depth understanding of pathogen-host interactions rely on technological advances. AREAS COVERED Rational vaccine development can be effectively approached by conceptual separation of, on one hand, design of immunogens from improving their presentation to the immune system and, on the other, induction of antibodies from induction of killer CD8(+) T cells. The biggest roadblock for many vaccines is the pathogens' variability. This is best tackled by focusing both antibodies and T cells on the functionally most conserved regions of proteins common to many variants, including escape mutants. For vectored vaccines, these 'universal' subunit immunogens are most efficiently delivered using heterologous prime-boost regimens, which can be further optimised by adjuvantation and route of delivery. EXPERT OPINION Development of vaccines against human diseases has many features in common. Acceleration of vaccine discovery depends on basic research and new technologies. Novel strategies should be safely, but rapidly tested in humans. While out-of-the-box thinking is important, vaccine success largely depends on incremental advances best achieved through small, systematic, iterative clinical studies. Failures are inevitable, but the end rewards are huge. The future will be exciting.
Collapse
Affiliation(s)
- Tomáš Hanke
- The Jenner Institute, University of Oxford , Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ , UK
| |
Collapse
|
339
|
Rieder F, Steininger C. Cytomegalovirus vaccine: phase II clinical trial results. Clin Microbiol Infect 2014; 20 Suppl 5:95-102. [PMID: 24283990 DOI: 10.1111/1469-0691.12449] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytomegalovirus (CMV) is one of the most significant viral pathogens during pregnancy and in immunocompromised patients. Antiviral prophylactic strategies are limited by toxicities, drug-drug interactions and development of antiviral resistance. A safe and protective vaccine against CMV is highly desirable in view of the potential positive impact on CMV-associated morbidity and mortality as well as healthcare costs. Unfortunately, this demand could not be met in the past four decades although development of a CMV vaccine has been ranked at the highest priority by the US Institute of Medicine. Multiple different vaccine candidates have been developed and evaluated in phase I clinical trials and few succeeded to phase II trials. Nevertheless, two different vaccines showed recently promising results in trials that studied healthy adults and immunocompromised solid-organ and bone-marrow transplant recipients, respectively. The gB/MF59 vaccine exhibited a vaccine efficacy of 50% in healthy, postpartum females. In transplant patients, gB/MF59 and the DNA vaccine TransVax both limited the periods of viraemia and consequently the need for antiviral treatment. The success of these trials is encouraging and will probably give new impetus to the development of an effective CMV vaccine. Sterilizing immunity may not be attainable in the near future and may not be necessary for a CMV vaccine to have a significant impact on health care as discussed in the present review.
Collapse
Affiliation(s)
- F Rieder
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
340
|
Roff SR, Noon-Song EN, Yamamoto JK. The Significance of Interferon-γ in HIV-1 Pathogenesis, Therapy, and Prophylaxis. Front Immunol 2014; 4:498. [PMID: 24454311 PMCID: PMC3888948 DOI: 10.3389/fimmu.2013.00498] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/17/2013] [Indexed: 12/24/2022] Open
Abstract
Interferon-γ (IFNγ) plays various roles in the pathogenesis of HIV/AIDS. In an HIV-1 infected individual, the production of IFNγ is detected as early as the acute phase and continually detected throughout the course of infection. Initially produced to clear the primary infection, IFNγ together with other inflammatory cytokines are involved in establishing a chronic immune activation that exacerbates clinical diseases associated with AIDS. Unlike Type 1 IFNs, IFNγ has no direct antiviral activity against HIV-1 in primary cultures, as supported by the in vivo findings of IFNγ therapy in infected subjects. Results from both in vitro and ex vivo studies show that IFNγ can instead enhance HIV-1 replication and its associated diseases, and therapies aimed at decreasing its production are under consideration. On the other hand, IFNγ has been shown to enhance cytotoxic T lymphocytes and NK cell activities against HIV-1 infected cells. These activities are important in controlling HIV-1 replication in an individual and will most likely play a role in the prophylaxis of an effective vaccine against HIV-1. Additionally, IFNγ has been used in combination with HIV-1 vaccine to augment antiviral immunity. Technological advancements have focused on using IFNγ as a biological marker to analyze the type(s) of immunity generated by candidate HIV vaccines and the levels of immunity restored by anti-retroviral drug therapies or novel immunotherapies. Hence, in addition to its valuable ancillary role as a biological marker for the development of effective HIV-1 prophylactic and therapeutic strategies, IFNγ has a vital role in promoting the pathogenesis of HIV.
Collapse
Affiliation(s)
- Shannon R. Roff
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Ezra N. Noon-Song
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Janet K. Yamamoto
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
341
|
Sylwester AW, Hansen SG, Picker LJ. Quantification of T cell Antigen-specific Memory Responses in Rhesus Macaques, Using Cytokine Flow Cytometry (CFC, also Known as ICS and ICCS): Analysis of Flow Data. Bio Protoc 2014; 4:e1109. [PMID: 28280751 DOI: 10.21769/bioprotoc.1109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
What was initially termed 'CFC' (Cytokine Flow Cytometry) is now more commonly known as 'ICS' (Intra Cellular Staining), or less commonly as 'ICCS' (Intra Cellular Cytokine Staining). The key innovations were use of an effective permeant (allowing intracellular staining), and a reagent to disrupt secretion (trapping cytokines, thereby enabling accumulation of detectable intracellular signal). Because not all researchers who use the technique are interested in cytokines, the 'ICS' term has gained favor, though 'CFC' will be used here. CFC is a test of cell function, exposing lymphocytes to antigen in culture, then measuring any cytokine responses elicited. Test cultures are processed so as to stain cells with monoclonal antibodies tagged with fluorescent markers, and to chemically fix the cells and decontaminate the samples, using paraformaldehyde. CFC provides the powers of flow cytometry, which includes bulk sampling and multi-parametric cross-correlation, to the analysis of antigen-specific memory responses. A researcher using CFC is able to phenotypically characterize cells cultured with test antigen, and for phenotypic subsets (e.g. CD4+ or CD8+ T cells) determine the % frequency producing cytokine above background level. In contrast to ELISPOT and Luminex methods, CFC can correlate production of multiple cytokines from particular, phenotypically-characterized cells. The CFC assay is useful for detecting that an individual has had an antigen exposure (as in population screenings), or for following the emergence and persistence of antigen memories (as in studies of vaccination, infections, or pathogenesis). In addition to quantifying the % frequency of antigen-responding cells, mean fluorescence intensity can be used to assess how much of a cytokine is generated within responding cells. With the technological advance of flow cytometry, a current user of CFC often has access to 11 fluorescent channels (or even 18), making it possible to either highly-characterize the phenotypes of antigen-responding cells, or else simultaneously quantify the responses according to many cytokines or activation markers. Powerful software like FlowJo (TreeStar) and SPICE (NIAID) can be used to analyse the data, and to do sophisticated multivariate analysis of cytokine responses. The method described here is customized for cells from Rhesus macaque monkeys, and the extensive annotating notes represent a decade of accumulated technical experience. The same scheme is readily applicable to other mammalian cells (e.g. human or mouse), though the exact antibody clones will differ according to host system. The basic method described here incubates 1 × 106 Lymphocytes in 1 ml tube culture with antigen and co-stimulatory antibodies in the presence of Brefeldin A, prior to staining and fixation. Note: This is the second part of a two-part procedure. Part one has the same initial title, but the subtitle "From Assay Set-up to Data Acquisition (Sylwester et al., 2014)". The Abstract and Historical Background is the same for both documents.
Collapse
Affiliation(s)
- Andrew W Sylwester
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, USA
| |
Collapse
|
342
|
Sylwester A, Hansen S, Picker L. Quantification of T Cell Antigen-specific Memory Responses in Rhesus Macaques, Using Cytokine Flow Cytometry (CFC, also Known as ICS and ICCS): from Assay Set-up to Data Acquisition. Bio Protoc 2014. [DOI: 10.21769/bioprotoc.1110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
343
|
Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol 2014; 14:24-35. [PMID: 24336101 PMCID: PMC4032067 DOI: 10.1038/nri3567] [Citation(s) in RCA: 629] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Memory T cells constitute the most abundant lymphocyte population in the body for the majority of a person's lifetime; however, our understanding of memory T cell generation, function and maintenance mainly derives from mouse studies, which cannot recapitulate the exposure to multiple pathogens that occurs over many decades in humans. In this Review, we discuss studies focused on human memory T cells that reveal key properties of these cells, including subset heterogeneity and diverse tissue residence in multiple mucosal and lymphoid tissue sites. We also review how the function and the adaptability of human memory T cells depend on spatial and temporal compartmentalization.
Collapse
Affiliation(s)
- Donna L Farber
- 1] Columbia Center for Translational Immunology and Department of Microbiology and Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, New York 10032, USA. [2] Department of Surgery, Columbia University Medical Center, 650 West 168th Street, BB1501, New York 10032, USA
| | - Naomi A Yudanin
- Columbia Center for Translational Immunology and Department of Microbiology and Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, New York 10032, USA
| | - Nicholas P Restifo
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
344
|
Beverley PCL, Sridhar S, Lalvani A, Tchilian EZ. Harnessing local and systemic immunity for vaccines against tuberculosis. Mucosal Immunol 2014; 7:20-6. [PMID: 24253104 DOI: 10.1038/mi.2013.99] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/09/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023]
Abstract
The lung is the portal of entry for Mycobacterium tuberculosis (Mtb) and animal experimental evidence indicates that local immune defense mechanisms are crucial for protective immunity. Immunization via the lower respiratory tract efficiently induces a dividing, activated, antigen-dependent, lung-resident, memory T-cell population, which is partly recoverable by bronchoalveolar lavage. These cells can inhibit the growth of Mtb in the lungs immediately after infection. Delivery of appropriate signals to the lung innate immune system is critical for induction of effective local immunity. In contrast after parenteral immunization, antigen-specific cells may be found in lung tissue but few are recoverable by lavage and inhibition of mycobacterial growth is delayed. Harnessing both local and systemic immunity can provide highly effective protection in animal models and the evidence suggests that taken in aggregate, multiple animal models may predict the success of novel vaccine strategies in humans.
Collapse
Affiliation(s)
- P C L Beverley
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S Sridhar
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - A Lalvani
- TB Research Unit, National Heart and Lung Institute, Imperial College, London, UK
| | - E Z Tchilian
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
345
|
Deletion of the human cytomegalovirus US17 gene increases the ratio of genomes per infectious unit and alters regulation of immune and endoplasmic reticulum stress response genes at early and late times after infection. J Virol 2013; 88:2168-82. [PMID: 24335296 DOI: 10.1128/jvi.02704-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Human cytomegalovirus (HCMV) employs numerous strategies to combat, subvert, or co-opt host immunity. One evolutionary strategy for this involves capture of a host gene and then its successive duplication and divergence, forming a family of genes, many of which have immunomodulatory activities. The HCMV US12 family consists of 10 tandemly arranged sequence-related genes in the unique short (US) region of the HCMV genome (US12 to US21). Each gene encodes a protein possessing seven predicted transmembrane domains, patches of sequence similarity with cellular G-protein-coupled receptors, and the Bax inhibitor 1 family of antiapoptotic proteins. We show that one member, US17, plays an important role during virion maturation. Microarray analysis of cells infected with a recombinant HCMV isolate with a US17 deletion (the ΔUS17 mutant virus) revealed blunted host innate and interferon responses at early times after infection (12 h postinfection [hpi]), a pattern opposite that previously seen in the absence of the immunomodulatory tegument protein pp65 (pUL83). Although the ΔUS17 mutant virus produced numbers of infectious particles in fibroblasts equal to the numbers produced by the parental virus, it produced >3-fold more genome-containing noninfectious viral particles and delivered increased amounts of pp65 to newly infected cells. These results suggest that US17 has evolved to control virion composition, to elicit an appropriately balanced host immune response. At later time points (96 hpi), ΔUS17 mutant-infected cells displayed aberrant expression of several host endoplasmic reticulum stress response genes and chaperones, some of which are important for the final stages of virion assembly and egress. Our results suggest that US17 modulates host pathways to enable production of virions that elicit an appropriately balanced host immune response.
Collapse
|
346
|
Karpenko LI, Bazhan SI, Antonets DV, Belyakov IM. Novel approaches in polyepitope T-cell vaccine development against HIV-1. Expert Rev Vaccines 2013; 13:155-73. [PMID: 24308576 DOI: 10.1586/14760584.2014.861748] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
RV144 clinical trial was modestly effective in preventing HIV infection. New alternative approaches are needed to design improved HIV-1 vaccines and their delivery strategies. One of these approaches is construction of synthetic polyepitope HIV-1 immunogen using protective T- and B-cell epitopes that can induce broadly neutralizing antibodies and responses of cytotoxic (CD8(+) CTL) and helpers (CD4(+) Th) T-lymphocytes. This approach seems to be promising for designing of new generation of vaccines against HIV-1, enables in theory to cope with HIV-1 antigenic variability, focuses immune responses on protective determinants and enables to exclude from the vaccine compound that can induce autoantibodies or antibodies enhancing HIV-1 infectivity. Herein, the authors will focus on construction and rational design of polyepitope T-cell HIV-1 immunogens and their delivery, including: advantages and disadvantages of existing T-cell epitope prediction methods; features of organization of polyepitope immunogens, which can generate high-level CD8(+) and CD4(+) T-lymphocyte responses; the strategies to optimize efficient processing, presentation and immunogenicity of polyepitope constructs; original software to design polyepitope immunogens; and delivery vectors as well as mucosal strategies of vaccination. This new knowledge may bring us a one step closer to developing an effective T-cell vaccine against HIV-1, other chronic viral infections and cancer.
Collapse
Affiliation(s)
- Larisa I Karpenko
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk region, 630559, Russia
| | | | | | | |
Collapse
|
347
|
Schleiss MR. Developing a Vaccine against Congenital Cytomegalovirus (CMV) Infection: What Have We Learned from Animal Models? Where Should We Go Next? Future Virol 2013; 8:1161-1182. [PMID: 24523827 DOI: 10.2217/fvl.13.106] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Congenital human cytomegalovirus (HCMV) infection can lead to long-term neurodevelopmental sequelae, including mental retardation and sensorineural hearing loss. Unfortunately, CMVs are highly adapted to their specific species, precluding the evaluation of HCMV vaccines in animal models prior to clinical trials. Several species-specific CMVs have been characterized and developed in models of pathogenesis and vaccine-mediated protection against disease. These include the murine CMV (MCMV), the porcine CMV (PCMV), the rhesus macaque CMV (RhCMV), the rat CMV (RCMV), and the guinea pig CMV (GPCMV). Because of the propensity of the GPCMV to cross the placenta, infecting the fetus in utero, it has emerged as a model of particular interest in studying vaccine-mediated protection of the fetus. In this paper, a review of these various models, with particular emphasis on the value of the model in the testing and evaluation of vaccines against congenital CMV, is provided. Recent exciting developments and advances in these various models are summarized, and recommendations offered for high-priority areas for future study.
Collapse
Affiliation(s)
- Mark R Schleiss
- University of Minnesota Medical School Center for Infectious Diseases and Microbiology Translational Research Department of Pediatrics Division of Pediatric Infectious Diseases and Immunology 2001 6 Street SE Minneapolis, MN 55455-3007
| |
Collapse
|
348
|
Developing Combined HIV Vaccine Strategies for a Functional Cure. Vaccines (Basel) 2013; 1:481-96. [PMID: 26344343 PMCID: PMC4494210 DOI: 10.3390/vaccines1040481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Accepted: 10/12/2013] [Indexed: 11/16/2022] Open
Abstract
Increasing numbers of HIV-infected individuals have access to potent antiretroviral drugs that control viral replication and decrease the risk of transmission. However, there is no cure for HIV and new strategies have to be developed to reach an eradication of the virus or a natural control of viral replication in the absence of drugs (functional cure). Therapeutic vaccines against HIV have been evaluated in many trials over the last 20 years and important knowledge has been gained from these trials. However, the major obstacle to HIV eradication is the persistence of latent proviral reservoirs. Different molecules are currently tested in ART-treated subjects to reactivate these latent reservoirs. Such anti-latency agents should be combined with a vaccination regimen in order to control or eradicate reactivated latently-infected cells. New in vitro assays should also be developed to assess the success of tested therapeutic vaccines by measuring the immune-mediated killing of replication-competent HIV reservoir cells. This review provides an overview of the current strategies to combine HIV vaccines with anti-latency agents that could act as adjuvant on the vaccine-induced immune response as well as new tools to assess the efficacy of these approaches.
Collapse
|
349
|
Resistance to infection, early and persistent suppression of simian immunodeficiency virus SIVmac251 viremia, and significant reduction of tissue viral burden after mucosal vaccination in female rhesus macaques. J Virol 2013; 88:212-24. [PMID: 24155376 DOI: 10.1128/jvi.02523-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The efficacy of oral, intestinal, nasal, and vaginal vaccinations with DNA simian immunodeficiency virus (SIV)/interleukin-2 (IL-2)/IL-15, SIV Gag/Pol/Env recombinant modified vaccinia virus Ankara (rMVA), and AT-2 SIVmac239 inactivated particles was compared in rhesus macaques after low-dose vaginal challenge with SIVmac251. Intestinal immunization provided better protection from infection, as a significantly greater median number of challenges was necessary in this group than in the others. Oral and nasal vaccinations provided the most significant control of disease progression. Fifty percent of the orally and nasally vaccinated animals suppressed viremia to undetectable levels, while this occurred to a significantly lower degree in intestinally and vaginally vaccinated animals and in controls. Viremia remained undetectable after CD8(+) T-cell depletion in seven vaccinated animals that had suppressed viremia after infection, and tissue analysis for SIV DNA and RNA was negative, a result consistent with a significant reduction of viral activity. Regardless of the route of vaccination, mucosal vaccinations prevented loss of CD4(+) central memory and CD4(+)/α4β7(+) T-cell populations and reduced immune activation to different degrees. None of the orally vaccinated animals and only one of the nasally vaccinated animals developed AIDS after 72 to 84 weeks of infection, when the trial was closed. The levels of anti-SIV gamma interferon-positive, CD4(+), and CD8(+) T cells at the time of first challenge inversely correlated with viremia and directly correlated with protection from infection and longer survival.
Collapse
|
350
|
Affiliation(s)
- Chris A Benedict
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA +1001 858 752 6732
| |
Collapse
|