1
|
Eslamizar L, Petrovas C, Leggat DJ, Furr K, Lifton ML, Levine G, Ma S, Fletez-Brant C, Hoyland W, Prabhakaran M, Narpala S, Boswell K, Yamamoto T, Liao HX, Pickup D, Ramsburg E, Sutherland L, McDermott A, Roederer M, Montefiori D, Koup RA, Haynes BF, Letvin NL, Santra S. Recombinant MVA-prime elicits neutralizing antibody responses by inducing antigen-specific B cells in the germinal center. NPJ Vaccines 2021; 6:15. [PMID: 33495459 PMCID: PMC7835239 DOI: 10.1038/s41541-020-00277-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/07/2020] [Indexed: 01/23/2023] Open
Abstract
The RV144 HIV-1 vaccine trial has been the only clinical trial to date that has shown any degree of efficacy and associated with the presence of vaccine-elicited HIV-1 envelope-specific binding antibody and CD4+ T-cell responses. This trial also showed that a vector-prime protein boost combined vaccine strategy was better than when used alone. Here we have studied three different priming vectors-plasmid DNA, recombinant MVA, and recombinant VSV, all encoding clade C transmitted/founder Env 1086 C gp140, for priming three groups of six non-human primates each, followed by a protein boost with adjuvanted 1086 C gp120 protein. Our data showed that MVA-priming favors the development of higher antibody binding titers and neutralizing activity compared with other vectors. Analyses of the draining lymph nodes revealed that MVA-prime induced increased germinal center reactivity characterized by higher frequencies of germinal center (PNAhi) B cells, higher frequencies of antigen-specific B-cell responses as well as an increased frequency of the highly differentiated (ICOShiCD150lo) Tfh-cell subset.
Collapse
Affiliation(s)
- Leila Eslamizar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Integrative Toxicology, Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., 175 Briar Ridge Road, Ridgefield, CT, 06877, USA
| | - Constantinos Petrovas
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA.
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| | | | - Kathryn Furr
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michelle L Lifton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gail Levine
- Foundation for the National Institutes of Health, Bethesda, MD, USA
| | - Steven Ma
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | | | | | | | | | | | | | - Hua-Xin Liao
- Foundation for the National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | - Norman L Letvin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sampa Santra
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Fernández-Escobar M, Baldanta S, Reyburn H, Guerra S. Use of functional genomics to understand replication deficient poxvirus-host interactions. Virus Res 2016; 216:1-15. [PMID: 26519757 DOI: 10.1016/j.virusres.2015.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 10/22/2022]
Abstract
High-throughput genomics technologies are currently being used to study a wide variety of viral infections, providing insight into which cellular genes and pathways are regulated after infection, and how these changes are related, or not, to efficient elimination of the pathogen. This article will focus on how gene expression studies of infections with non-replicative poxviruses currently used as vaccine vectors provide a global perspective of the molecular events associated with the viral infection in human cells. These high-throughput genomics approaches have the potential to lead to the identification of specific new properties of the viral vector or novel cellular targets that may aid in the development of more effective pox-derived vaccines and antivirals.
Collapse
Affiliation(s)
- Mercedes Fernández-Escobar
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma, E-28029 Madrid, Spain
| | - Sara Baldanta
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma, E-28029 Madrid, Spain
| | - Hugh Reyburn
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma, E-28029 Madrid, Spain.
| |
Collapse
|
3
|
Prime-boost vaccine strategy against viral infections: Mechanisms and benefits. Vaccine 2016; 34:413-423. [DOI: 10.1016/j.vaccine.2015.11.062] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/01/2023]
|
4
|
Goulding J, Tahiliani V, Salek-Ardakani S. OX40:OX40L axis: emerging targets for improving poxvirus-based CD8(+) T-cell vaccines against respiratory viruses. Immunol Rev 2012; 244:149-68. [PMID: 22017437 PMCID: PMC3422077 DOI: 10.1111/j.1600-065x.2011.01062.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The human respiratory tract is an entry point for over 200 known viruses that collectively contribute to millions of annual deaths worldwide. Consequently, the World Health Organization has designated respiratory viral infections as a priority for vaccine development. Despite enormous advances in understanding the attributes of a protective mucosal antiviral immune response, current vaccines continue to fail in effectively generating long-lived protective CD8(+) T-cell immunity. To date, the majority of licensed human vaccines afford protection against infectious pathogens through the generation of specific immunoglobulin responses. In recent years, the selective manipulation of specific costimulatory pathways, which are critical in regulating T cell-mediated immune responses, has generated increasing interest. Impressive results in animal models have shown that the tumor necrosis factor receptor (TNFR) family member OX40 (CD134) and its binding partner OX40L (CD252) are key costimulatory molecules involved in the generation of protective CD8(+) T-cell responses at mucosal surfaces, such as the lung. In this review, we highlight these new findings with a particular emphasis on their potential as immunological adjuvants to enhance poxvirus-based CD8(+) T-cell vaccines.
Collapse
Affiliation(s)
- John Goulding
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, San Diego, CA, USA
| | | | | |
Collapse
|
5
|
Elnekave M, Furmanov K, Hovav AH. Intradermal naked plasmid DNA immunization: mechanisms of action. Expert Rev Vaccines 2012; 10:1169-82. [PMID: 21854310 DOI: 10.1586/erv.11.66] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Plasmid DNA is a promising vaccine modality that is regularly examined in prime-boost immunization regimens. Recent advances in skin immunity increased our understanding of the sophisticated cutaneous immune network, which revived scientific interest in delivering vaccines to the skin. Intradermal administration of plasmid DNA via needle injection is a simple and inexpensive procedure that exposes the plasmid and its encoded antigen to the dermal immune surveillance system. This triggers unique mechanisms for eliciting local and systemic immunity that can confer protection against pathogens and tumors. Understanding the mechanisms of intradermal plasmid DNA immunization is essential for enhancing and modulating its immunogenicity. With regard to vaccination, this is of greater importance as this routine injection technique is highly desirable for worldwide immunization. This article will focus on the current understanding of the mechanisms involved in antigen expression and presentation during primary and secondary syringe and needle intradermal plasmid DNA immunization.
Collapse
Affiliation(s)
- Mazal Elnekave
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine, PO Box 122722, Jerusalem 91120, Israel
| | | | | |
Collapse
|
6
|
Immunization with HIV Gag targeted to dendritic cells followed by recombinant New York vaccinia virus induces robust T-cell immunity in nonhuman primates. Proc Natl Acad Sci U S A 2011; 108:7131-6. [PMID: 21467219 DOI: 10.1073/pnas.1103869108] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protein vaccines, if rendered immunogenic, would facilitate vaccine development against HIV and other pathogens. We compared in nonhuman primates (NHPs) immune responses to HIV Gag p24 within 3G9 antibody to DEC205 ("DEC-HIV Gag p24"), an uptake receptor on dendritic cells, to nontargeted protein, with or without poly ICLC, a synthetic double stranded RNA, as adjuvant. Priming s.c. with 60 μg of both HIV Gag p24 vaccines elicited potent CD4(+) T cells secreting IL-2, IFN-γ, and TNF-α, which also proliferated. The responses increased with each of three immunizations and recognized multiple Gag peptides. DEC-HIV Gag p24 showed better cross-priming for CD8(+) T cells, whereas the avidity of anti-Gag antibodies was ∼10-fold higher with nontargeted Gag 24 protein. For both protein vaccines, poly ICLC was essential for T- and B-cell immunity. To determine whether adaptive responses could be further enhanced, animals were boosted with New York vaccinia virus (NYVAC)-HIV Gag/Pol/Nef. Gag-specific CD4(+) and CD8(+) T-cell responses increased markedly after priming with both protein vaccines and poly ICLC. These data reveal qualitative differences in antibody and T-cell responses to DEC-HIV Gag p24 and Gag p24 protein and show that prime boost with protein and adjuvant followed by NYVAC elicits potent cellular immunity.
Collapse
|
7
|
Wilks AB, Christian EC, Seaman MS, Sircar P, Carville A, Gomez CE, Esteban M, Pantaleo G, Barouch DH, Letvin NL, Permar SR. Robust vaccine-elicited cellular immune responses in breast milk following systemic simian immunodeficiency virus DNA prime and live virus vector boost vaccination of lactating rhesus monkeys. THE JOURNAL OF IMMUNOLOGY 2010; 185:7097-106. [PMID: 21041730 DOI: 10.4049/jimmunol.1002751] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Breast milk transmission of HIV remains an important mode of infant HIV acquisition. Enhancement of mucosal HIV-specific immune responses in milk of HIV-infected mothers through vaccination may reduce milk virus load or protect against virus transmission in the infant gastrointestinal tract. However, the ability of HIV/SIV strategies to induce virus-specific immune responses in milk has not been studied. In this study, five uninfected, hormone-induced lactating, Mamu A*01(+) female rhesus monkey were systemically primed and boosted with rDNA and the attenuated poxvirus vector, NYVAC, containing the SIVmac239 gag-pol and envelope genes. The monkeys were boosted a second time with a recombinant Adenovirus serotype 5 vector containing matching immunogens. The vaccine-elicited immunodominant epitope-specific CD8(+) T lymphocyte response in milk was of similar or greater magnitude than that in blood and the vaginal tract but higher than that in the colon. Furthermore, the vaccine-elicited SIV Gag-specific CD4(+) and CD8(+) T lymphocyte polyfunctional cytokine responses were more robust in milk than in blood after each virus vector boost. Finally, SIV envelope-specific IgG responses were detected in milk of all monkeys after vaccination, whereas an SIV envelope-specific IgA response was only detected in one vaccinated monkey. Importantly, only limited and transient increases in the proportion of activated or CCR5-expressing CD4(+) T lymphocytes in milk occurred after vaccination. Therefore, systemic DNA prime and virus vector boost of lactating rhesus monkeys elicits potent virus-specific cellular and humoral immune responses in milk and may warrant further investigation as a strategy to impede breast milk transmission of HIV.
Collapse
Affiliation(s)
- Andrew B Wilks
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Kim D, Hung CF, Wu TC, Park YM. DNA vaccine with α-galactosylceramide at prime phase enhances anti-tumor immunity after boosting with antigen-expressing dendritic cells. Vaccine 2010; 28:7297-305. [PMID: 20817010 DOI: 10.1016/j.vaccine.2010.08.079] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 07/22/2010] [Accepted: 08/19/2010] [Indexed: 10/19/2022]
Abstract
DNA vaccines contribute to a promising new approach for the generation of cytotoxic T lymphocytes (CTL). DNA vaccines do have several disadvantages, including poor immunogenicity and oncogene expression. We used the natural killer T-cell (NKT) ligand α-galactosylceramide (α-GalCer) as an adjuvant to prime initial DNA vaccination; and used the potent immune-stimulatory tumor antigen-expressing dendritic cells (DCs) as a booster vaccination. A DNA vaccine expressing human papillomavirus (HPV) type 16 E7 (pcDNA3-CRT/E7) was combined with α-GalCer at the prime phase, and generated a higher number of E7-specific CD8(+) T-cells in vaccinated mice than vaccine used at boost phase. Therefore, priming with a DNA vaccine in the presence of α-GalCer and boosting with E7-pulsed DC-1 led to a significant enhancement of E7-specific CD8(+) effector and memory T-cells as well as significantly improved therapeutic and preventive effects against an E7-expressing tumor model (TC-1) in vaccinated mice. Our findings suggested that the potency of a DNA vaccine combined with α-GalCer could be further enhanced by boosting with an antigen-expressing DC-based vaccine to generate anti-tumor immunity.
Collapse
Affiliation(s)
- Daejin Kim
- Department of Anatomy, Chung-Ang University, College of Medicine, Seoul, Republic of Korea.
| | | | | | | |
Collapse
|
9
|
Sun Y, Santra S, Buzby AP, Mascola JR, Nabel GJ, Letvin NL. Recombinant vector-induced HIV/SIV-specific CD4+ T lymphocyte responses in rhesus monkeys. Virology 2010; 406:48-55. [PMID: 20667574 DOI: 10.1016/j.virol.2010.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 06/14/2010] [Accepted: 07/02/2010] [Indexed: 11/25/2022]
Abstract
The recently reported modest success of the RV144 Thai trial vaccine regimen in preventing HIV-1 acquisition has focused interest on the potential contribution to that protection of vaccine-elicited CD4(+) T cell responses. We evaluated the induction of virus-specific CD4(+) T cell responses in rhesus monkeys using a series of diverse vaccine vectors. We assessed both the magnitudes and functional profiles of the antigen-specific CD4(+) T cells by measuring cytokine production, memory differentiation, and the expression of mucosal homing molecules. We found that DNA prime/recombinant MVA boost immunizations induced particularly high-frequency virus-specific CD4(+) T cell responses with polyfunctional repertoires, and these responses were partially preserved following SHIV-89.6P challenge. The majority of the vaccine-elicited CD4(+) T cells were CD28(+) memory T cells that expressed low levels of beta7. Neither the magnitudes nor the functional profiles of the virus-specific CD4(+) T cells generated by vaccination were associated with a preservation of CD4(+) T cells or control of viral replication following SHIV-89.6P challenge. Interestingly, monkeys primed with recombinant Ad5 immunogens showed a dramatic expansion of both the magnitude and polyfunctionality of the vaccine-elicited CD4(+) T cell responses following envelope protein boost. These results demonstrate that vaccine strategies that include recombinant MVA or recombinant Ad5 vectors can elicit robust CD4(+) T cell responses.
Collapse
Affiliation(s)
- Yue Sun
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
10
|
Mosaic vaccines elicit CD8+ T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys. Nat Med 2010; 16:324-8. [PMID: 20173754 PMCID: PMC2834806 DOI: 10.1038/nm.2108] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 01/27/2010] [Indexed: 11/14/2022]
Abstract
An effective HIV vaccine must elicit immune responses that recognize genetically diverse viruses1, 2. It must generate CD8+ T lymphocytes that control HIV replication and CD4+ T lymphocytes that provide help for the generation and maintenance of both cellular and humoral immune responses against the virus3–5. Creating immunogens that can elicit cellular immune responses against the genetically varied circulating isolates of HIV presents an important challenge for creating an AIDS vaccine6, 7. Polyvalent mosaic immunogens derived by in silico recombination of natural strains of HIV are designed to induce cellular immune responses that recognize genetically diverse circulating virus isolates8. In the present study we immunized rhesus monkeys by plasmid DNA prime/ recombinant vaccinia virus boost using vaccine constructs expressing either consensus or polyvalent mosaic proteins. The mosaic immunogens elicited CD8+ T lymphocyte responses to more epitopes of each viral protein than the consensus immunogens, and to more variant sequences of CD8+ T lymphocyte epitopes. This increased breadth and depth of epitope recognition may contribute both to protection against infection by genetically diverse viruses and to the control of variant viruses that emerge as they mutate away from recognition by cytotoxic T lymphocytes.
Collapse
|
11
|
Isakov D, Dzutsev A, Belyakov IM, Berzofsky JA. Non-equilibrium and differential function between intraepithelial and lamina propria virus-specific TCRalphabeta(+) CD8alphabeta(+) T cells in the small intestinal mucosa. Mucosal Immunol 2009; 2:450-61. [PMID: 19571797 PMCID: PMC3224999 DOI: 10.1038/mi.2009.95] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The gastrointestinal mucosa regularly encounters commensal and pathogenic microbiota. Gut mucosal lymphocytes consist of two phenotypically different populations residing in the intestinal intraepithelial (IEL) compartment and lamina propria (LP). Little is known about compositional and functional differences of antigen-specific T cells from these mucosal compartments after mucosal infection, or the degree of trafficking between them. We here studied the B8R(20-27)-specific CD8 T-cell response in LP and IEL compartments after intrarectal immunization with modified vaccinia virus Ankara (MVA). CD8(+) T cells in the IEL compartment had much lower avidity than in the LP or spleen during acute and memory phases. Surprisingly, the TCR Vbeta-chain distribution of antigen-specific T cells and the length of the CDR3 region of the dominant Vbeta genes showed substantial dissimilarities between IEL and LP antigen-specific CD8alphabeta T cells in individual mice, increasing with time. We show functional and compositional differences between these mucosal compartments during the effector and memory phases of the immune response, indicating limited crosstalk and microenvironmental differences between the IEL, LP, and spleen. The restricted migration of cells from each of these mucosal compartments could partly account for a founder effect we observed in the IEL TCRalphabeta CD8alphabeta epitope-specific repertoire that might impact protective efficacy.
Collapse
Affiliation(s)
- D Isakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - A Dzutsev
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - IM Belyakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA, Midwest Research Institute, Frederick, Maryland, USA
| | - JA Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
12
|
Lu Y, Ouyang K, Fang J, Zhang H, Wu G, Ma Y, Zhang Y, Hu X, Jin L, Cao R, Fan H, Li T, Liu J. Improved efficacy of DNA vaccination against prostate carcinoma by boosting with recombinant protein vaccine and by introduction of a novel adjuvant epitope. Vaccine 2009; 27:5411-8. [PMID: 19616501 DOI: 10.1016/j.vaccine.2009.06.089] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 06/01/2009] [Accepted: 06/09/2009] [Indexed: 11/24/2022]
Abstract
DNA vaccine represents an attractive approach for cancer treatment by inducing active immune-deprivation of gastrin-releasing peptide (GRP) from tumor cells, the growth of which is dependent on the stimulation of GRP. In this study, we developed a DNA vaccine using a plasmid vector to deliver the immunogen of six copies of the B cell epitope GRP(18-27) (GRP6). In order to increase the potency of this DNA vaccine, multiple strategies have been applied including DNA-prime protein-boost immunization and introduction of a foreign T-helper epitope into DNA vaccine. Mice vaccinated DNA vaccine boosting with HSP65-GRP6 protein induced high titer and relatively high avidity of anti-GRP antibodies as well as inhibition effect on the growth of murine prostate carcinoma, superior to the treatment using DNA alone or BCG priming HSP65-GRP6 protein boosting. Furthermore, the introduction of a novel foreign T-helper epitope into the GRP DNA vaccine showed a markedly stronger humoral immune response against GRP and tumor rejection even than the DNA-prime protein-boost strategy. No further stronger immunogenicity of this foreign T-helper epitope modified DNA vaccine was observed even using the strategy of modified DNA vaccine-priming and HSP65-GRP6 boosting method. The data presented demonstrate that improvement of potency of anti-GRP DNA vaccine with the above two feasible approaches should offer useful methods in the development of new DNA vaccine against growth factors for cancer immunotherapy.
Collapse
Affiliation(s)
- Yong Lu
- Minigene Pharmacy Laboratory, Biopharmaceutical College, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Immunogenicity of recombinant Modified Vaccinia Ankara following a single or multi-dose vaccine regimen in rhesus monkeys. Vaccine 2009; 27:1549-56. [PMID: 19168105 DOI: 10.1016/j.vaccine.2009.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 12/23/2008] [Accepted: 01/05/2009] [Indexed: 11/24/2022]
Abstract
Modified Vaccinia Ankara (MVA) is a replication-defective strain of vaccinia virus (VV) that is being investigated in humans as an alternative vaccine against smallpox. Understanding the parameters of a MVA vaccine regimen that can effectively enhance protective immunity will be important for clinical development. The present studies utilize cohorts of rhesus monkeys immunized with recombinant MVA (rMVA) or recombinant VV (rVV) vaccine vectors to investigate the magnitude, breadth, and durability of anti-VV immunity elicited by a single or multi-dose vaccine regimen. These data demonstrate that a single immunization with rMVA elicits weaker cellular and humoral immunity compared to a single inoculation with rVV. However, vaccine-elicited antibody responses, but not T cell responses, are significantly enhanced with repeated immunizations of rMVA. Importantly, only monkeys receiving up to four inoculations with rMVA generated neutralizing antibody (NAb) responses that were comparable in magnitude and durability to those elicited in monkeys receiving two inoculations with rVV. These data also show that the breadth of antibody responses against protein antigens associated with two antigenically distinct forms of infectious VV are similar in rMVA- and rVV-immunized monkeys. Together, these studies suggest that a multi-dose vaccine regimen utilizing up to four inoculations of MVA generates robust and durable antibody-mediated immunity comparable to that elicited by replication-competent VV.
Collapse
|
14
|
Reynolds MR, Weiler AM, Weisgrau KL, Piaskowski SM, Furlott JR, Weinfurter JT, Kaizu M, Soma T, León EJ, MacNair C, Leaman DP, Zwick MB, Gostick E, Musani SK, Price DA, Friedrich TC, Rakasz EG, Wilson NA, McDermott AB, Boyle R, Allison DB, Burton DR, Koff WC, Watkins DI. Macaques vaccinated with live-attenuated SIV control replication of heterologous virus. ACTA ACUST UNITED AC 2008; 205:2537-50. [PMID: 18838548 PMCID: PMC2571929 DOI: 10.1084/jem.20081524] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
An effective AIDS vaccine will need to protect against globally diverse isolates of HIV. To address this issue in macaques, we administered a live-attenuated simian immunodeficiency virus (SIV) vaccine and challenged with a highly pathogenic heterologous isolate. Vaccinees reduced viral replication by ∼2 logs between weeks 2–32 (P ≤ 0.049) postchallenge. Remarkably, vaccinees expressing MHC-I (MHC class I) alleles previously associated with viral control completely suppressed acute phase replication of the challenge virus, implicating CD8+ T cells in this control. Furthermore, transient depletion of peripheral CD8+ lymphocytes in four vaccinees during the chronic phase resulted in an increase in virus replication. In two of these animals, the recrudescent virus population contained only the vaccine strain and not the challenge virus. Alarmingly, however, we found evidence of recombinant viruses emerging in some of the vaccinated animals. This finding argues strongly against an attenuated virus vaccine as a solution to the AIDS epidemic. On a more positive note, our results suggest that MHC-I–restricted CD8+ T cells contribute to the protection induced by the live-attenuated SIV vaccine and demonstrate that vaccine-induced CD8+ T cell responses can control replication of heterologous challenge viruses.
Collapse
Affiliation(s)
- Matthew R Reynolds
- AIDS Vaccine Research Laboratory, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Magnitude and quality of vaccine-elicited T-cell responses in the control of immunodeficiency virus replication in rhesus monkeys. J Virol 2008; 82:8812-9. [PMID: 18579590 DOI: 10.1128/jvi.00204-08] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
While a diversity of immunogens that elicit qualitatively different cellular immune responses are being assessed in clinical human immunodeficiency virus vaccine trials, the consequences of those varied responses for viral control remain poorly understood. In the present study, we evaluated the induction of virus-specific T-cell responses in rhesus monkeys using a series of diverse vaccine vectors. We assessed both the magnitude and the functional profile of the virus-specific CD8(+) T cells by measuring gamma interferon, interleukin-2, and tumor necrosis factor alpha production. We found that the different vectors generated virus-specific T-cell responses of different magnitudes and with different functional profiles. Heterologous prime-boost vaccine regimens induced particularly high-frequency virus-specific T-cell responses with polyfunctional repertoires. Yet, immediately after a pathogenic simian-human immunodeficiency virus (SHIV) challenge, no significant differences were observed between these cohorts of vaccinated monkeys in the magnitudes or the functional profiles of their virus-specific CD8(+) T cells. This finding suggests that the high viral load shapes the functional repertoire of the cellular immune response during primary infection. Nevertheless, in all vaccination regimens, higher frequency and more polyfunctional vaccine-elicited virus-specific CD8(+) T-cell responses were associated with better viral control after SHIV challenge. These observations highlight the contributions of both the quality and the magnitude of vaccine-elicited cellular immune responses in the control of immunodeficiency virus replication.
Collapse
|
16
|
Meyers A, Chakauya E, Shephard E, Tanzer FL, Maclean J, Lynch A, Williamson AL, Rybicki EP. Expression of HIV-1 antigens in plants as potential subunit vaccines. BMC Biotechnol 2008; 8:53. [PMID: 18573204 PMCID: PMC2443125 DOI: 10.1186/1472-6750-8-53] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 06/23/2008] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) has infected more than 40 million people worldwide, mainly in sub-Saharan Africa. The high prevalence of HIV-1 subtype C in southern Africa necessitates the development of cheap, effective vaccines. One means of production is the use of plants, for which a number of different techniques have been successfully developed. HIV-1 Pr55Gag is a promising HIV-1 vaccine candidate: we compared the expression of this and a truncated Gag (p17/p24) and the p24 capsid subunit in Nicotiana spp. using transgenic plants and transient expression via Agrobacterium tumefaciens and recombinant tobamovirus vectors. We also investigated the influence of subcellular localisation of recombinant protein to the chloroplast and the endoplasmic reticulum (ER) on protein yield. We partially purified a selected vaccine candidate and tested its stimulation of a humoral and cellular immune response in mice. RESULTS Both transient and transgenic expression of the HIV antigens were successful, although expression of Pr55Gag was low in all systems; however, the Agrobacterium-mediated transient expression of p24 and p17/p24 yielded best, to more than 1 mg p24/kg fresh weight. Chloroplast targeted protein levels were highest in transient and transgenic expression of p24 and p17/p24. The transiently-expressed p17/p24 was not immunogenic in mice as a homologous vaccine, but it significantly boosted a humoral and T cell immune response primed by a gag DNA vaccine, pTHGagC. CONCLUSION Transient agroinfiltration was best for expression of all of the recombinant proteins tested, and p24 and p17/p24 were expressed at much higher levels than Pr55Gag. Our results highlight the usefulness of plastid signal peptides in enhancing the production of recombinant proteins meant for use as vaccines. The p17/p24 protein effectively boosted T cell and humoral responses in mice primed by the DNA vaccine pTHGagC, showing that this plant-produced protein has potential for use as a vaccine.
Collapse
MESH Headings
- AIDS Vaccines/biosynthesis
- AIDS Vaccines/genetics
- Adjuvants, Immunologic/genetics
- Agrobacterium tumefaciens/genetics
- Animals
- Chloroplasts/genetics
- Chloroplasts/metabolism
- Endoplasmic Reticulum/genetics
- Endoplasmic Reticulum/metabolism
- Female
- Gene Expression
- Gene Expression Regulation, Plant
- Genes, gag
- Genetic Vectors
- HIV Antigens/biosynthesis
- HIV Antigens/genetics
- HIV Antigens/immunology
- HIV Infections/immunology
- HIV Seronegativity
- HIV-1/genetics
- Humans
- Mice
- Mice, Inbred BALB C
- Plants, Genetically Modified
- Nicotiana/genetics
- Tobamovirus/genetics
- Transformation, Genetic
- Vaccines, Subunit/biosynthesis
- Vaccines, Subunit/genetics
- gag Gene Products, Human Immunodeficiency Virus/biosynthesis
- gag Gene Products, Human Immunodeficiency Virus/genetics
Collapse
Affiliation(s)
- Ann Meyers
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Ereck Chakauya
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
- CSIR Biosciences, Pretoria 0001, South Africa
| | - Enid Shephard
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- MRC/UCT Liver Research Centre, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Fiona L Tanzer
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - James Maclean
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Alisson Lynch
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Observatory 7925, South Africa
| | - Edward P Rybicki
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| |
Collapse
|
17
|
Modifying the HIV-1 env gp160 gene to improve pDNA vaccine-elicited cell-mediated immune responses. Vaccine 2008; 26:5083-94. [PMID: 18485543 DOI: 10.1016/j.vaccine.2008.03.092] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Plasmid DNA (pDNA) vaccines are effective at eliciting immune responses in a wide variety of animal model systems, however, pDNA vaccines have generally been incapable of inducing robust immune responses in clinical trials. Therefore, to identify means to improve pDNA vaccine performance, we compared various post-transcriptional and post-translational genetic modifications for their ability to improve antigen-specific CMI responses. Mice vaccinated using a sub-optimal 100 mcg dose of a pDNA encoding an unmodified primary isolate HIV-1(6101) env gp160 failed to demonstrate measurable env-specific CMI responses. In contrast, significant env-specific CMI responses were seen in mice immunized with pDNA expression vectors encoding env genes modified by RNA optimization or codon optimization. Further modification of the RNA optimized env gp160 gene by the addition of (i) a simian retrovirus type 1 constitutive RNA transport element; (ii) a murine intracisternal A-particle derived RNA transport element; (iii) a tissue plasminogen activator protein signal leader sequences; (iv) a beta-catenin derived ubiquitination target sequence; or (v) a monocyte chemotactic protein-3 derived signal sequence failed to further improve the induction of env-specific CMI responses. Therefore, modification of the env gp160 gene by RNA or codon optimization alone is necessary for high-level rev-independent expression and results in robust env-specific CMI responses in immunized mice. Importantly, further modification(s) of the env gene to alter cellular localization or increase proteolytic processing failed to result in increased env-specific immune responses. These results have important implications for the design and development of an efficacious vaccine for the prevention of HIV-1 infection.
Collapse
|
18
|
Halsey RJ, Tanzer FL, Meyers A, Pillay S, Lynch A, Shephard E, Williamson AL, Rybicki EP. Chimaeric HIV-1 subtype C Gag molecules with large in-frame C-terminal polypeptide fusions form virus-like particles. Virus Res 2008; 133:259-68. [PMID: 18329748 DOI: 10.1016/j.virusres.2008.01.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 01/16/2008] [Accepted: 01/21/2008] [Indexed: 10/22/2022]
Abstract
HIV-1 Pr55 Gag virus-like particles (VLPs) are strong immunogens with potential as candidate HIV vaccines. VLP immunogenicity can be broadened by making chimaeric Gag molecules: however, VLPs incorporating polypeptides longer than 200 aa fused in frame with Gag have not yet been reported. We constructed a range of gag-derived genes encoding in-frame C-terminal fusions of myristoylation-competent native Pr55Gag and p6-truncated Gag (Pr50Gag) to test the effects of polypeptide length and sequence on VLP formation and morphology, in an insect cell expression system. Fused sequences included a modified reverse transcriptase-Tat-Nef fusion polypeptide (RTTN, 778 aa), and truncated versions of RTTN ranging from 113 aa to 450 aa. Baculovirus-expressed chimaeric proteins were examined by western blot and electron microscopy. All chimaeras formed VLPs which could be purified by sucrose gradient centrifugation. VLP diameter increased with protein MW, from approximately 100 nm for Pr55Gag to approximately 250 nm for GagRTTN. The presence or absence of the Gag p6 region did not obviously affect VLP formation or appearance. GagRT chimaeric particles were successfully used in mice to boost T-cell responses to Gag and RT that were elicited by a DNA vaccine encoding a GagRTTN polypeptide, indicating the potential of such chimaeras to be used as candidate HIV vaccines.
Collapse
Affiliation(s)
- Richard J Halsey
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, PO Observatory 7925, Cape Town, South Africa
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Mooij P, Balla-Jhagjhoorsingh SS, Koopman G, Beenhakker N, van Haaften P, Baak I, Nieuwenhuis IG, Kondova I, Wagner R, Wolf H, Gómez CE, Nájera JL, Jiménez V, Esteban M, Heeney JL. Differential CD4+ versus CD8+ T-cell responses elicited by different poxvirus-based human immunodeficiency virus type 1 vaccine candidates provide comparable efficacies in primates. J Virol 2008; 82:2975-88. [PMID: 18184713 PMCID: PMC2258966 DOI: 10.1128/jvi.02216-07] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Accepted: 12/13/2007] [Indexed: 12/20/2022] Open
Abstract
Poxvirus vectors have proven to be highly effective for boosting immune responses in diverse vaccine settings. Recent reports reveal marked differences in the gene expression of human dendritic cells infected with two leading poxvirus-based human immunodeficiency virus (HIV) vaccine candidates, New York vaccinia virus (NYVAC) and modified vaccinia virus Ankara (MVA). To understand how complex genomic changes in these two vaccine vectors translate into antigen-specific systemic immune responses, we undertook a head-to-head vaccine immunogenicity and efficacy study in the pathogenic HIV type 1 (HIV-1) model of AIDS in Indian rhesus macaques. Differences in the immune responses in outbred animals were not distinguished by enzyme-linked immunospot assays, but differences were distinguished by multiparameter fluorescence-activated cell sorter analysis, revealing a difference between the number of animals with both CD4(+) and CD8(+) T-cell responses to vaccine inserts (MVA) and those that elicit a dominant CD4(+) T-cell response (NYVAC). Remarkably, vector-induced differences in CD4(+)/CD8(+) T-cell immune responses persisted for more than a year after challenge and even accompanied antigenic modulation throughout the control of chronic infection. Importantly, strong preexposure HIV-1/simian immunodeficiency virus-specific CD4(+) T-cell responses did not prove deleterious with respect to accelerated disease progression. In contrast, in this setting, animals with strong vaccine-induced polyfunctional CD4(+) T-cell responses showed efficacies similar to those with stronger CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Petra Mooij
- Department of Virology, Biomedical Primate Research Centre (BPRC), P.O. Box 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Sekaly RP. The failed HIV Merck vaccine study: a step back or a launching point for future vaccine development? ACTA ACUST UNITED AC 2008; 205:7-12. [PMID: 18195078 PMCID: PMC2234358 DOI: 10.1084/jem.20072681] [Citation(s) in RCA: 307] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The world of human immunodeficiency virus (HIV) vaccines has suffered a baffling setback. The first trial of a vaccine designed to elicit strong cellular immunity has shown no protection against infection. More alarmingly, the vaccine appeared to increase the rate of HIV infection in individuals with prior immunity against the adenovirus vector used in the vaccine. A new study in this issue suggests that a different vaccine approach—using a DNA prime/poxvirus boost strategy—induces polyfunctional immune responses to an HIV immunogen. The disappointing results of the recent vaccine trial suggest that a more thorough assessment of vaccine-induced immune responses is urgently needed, and that more emphasis should be placed on primate models before efficacy trials are undertaken.
Collapse
Affiliation(s)
- Rafick-Pierre Sekaly
- Université de Montréal, CR-CHUM, Institut National de la Santé et de la Recherche Médicale U743, Montréal, Québec H2X1P1, Canada.
| |
Collapse
|
21
|
Clonal focusing of epitope-specific CD8+ T lymphocytes in rhesus monkeys following vaccination and simian-human immunodeficiency virus challenge. J Virol 2007; 82:805-16. [PMID: 17977967 DOI: 10.1128/jvi.01038-07] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
To afford the greatest possible immune protection, candidate human immunodeficiency virus (HIV) vaccines must generate diverse and long-lasting CD8(+) T lymphocyte responses. In the present study, we evaluate T-cell receptor Vbeta (variable region beta) gene usage and a CDR3 (complementarity-determining region 3) sequence to assess the clonality of epitope-specific CD8(+) T lymphocytes generated in rhesus monkeys following vaccination and simian-human immunodeficiency virus (SHIV) challenge. We found that vaccine-elicited epitope-specific CD8(+) T lymphocytes have a clonal diversity comparable to those cells generated in response to SHIV infection. Moreover, we show that the clonal diversity of vaccine-elicited CD8(+) T-lymphocyte responses is dictated by the epitope sequence and is not affected by the mode of antigen delivery to the immune system. Clonal CD8(+) T-lymphocyte populations persisted following boosting with different vectors, and these clonal cell populations could be detected for as long as 4 years after SHIV challenge. Finally, we show that the breadth of these epitope-specific T lymphocytes transiently focuses in response to intense SHIV replication. These observations demonstrate the importance of the initial immune response to SHIV, induced by vaccination or generated during primary infection, in determining the clonal diversity of cell-mediated immune responses and highlight the focusing of this clonal diversity in the setting of high viral loads. Circumventing this restricted CD8(+) T-lymphocyte clonal diversity may present a significant challenge in the development of an effective HIV vaccine strategy.
Collapse
|
22
|
Seaman MS, Leblanc DF, Grandpre LE, Bartman MT, Montefiori DC, Letvin NL, Mascola JR. Standardized assessment of NAb responses elicited in rhesus monkeys immunized with single- or multi-clade HIV-1 envelope immunogens. Virology 2007; 367:175-86. [PMID: 17599382 PMCID: PMC2075526 DOI: 10.1016/j.virol.2007.05.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 04/30/2007] [Accepted: 05/21/2007] [Indexed: 11/26/2022]
Abstract
The genetic diversity of HIV-1 envelope glycoproteins (Env) remains a major obstacle to the development of an antibody-based AIDS vaccine. The present studies examine the breadth and magnitude of neutralizing antibody (NAb) responses in rhesus monkeys after immunization with DNA prime-recombinant adenovirus (rAd) boost vaccines encoding either single or multiple genetically distant Env immunogens, and subsequently challenged with a pathogenic simian-human immunodeficiency virus (SHIV-89.6P). Using a standardized multi-tier panel of reference Env pseudoviruses for NAb assessment, we show that monkeys immunized with a mixture of Env immunogens (clades A, B, and C) exhibited a greater breadth of NAb activity against neutralization-sensitive Tier 1 viruses following both vaccination and challenge compared to monkeys immunized with a single Env immunogen (clade B or C). However, all groups of Env-vaccinated monkeys demonstrated only limited neutralizing activity against Tier 2 pseudoviruses, which are more characteristic of the neutralization sensitivity of circulating HIV-1. Notably, the development of a post-challenge NAb response against SHIV-89.6P was similar in monkeys receiving either clade B, clade C, or clade A+B+C Env immunogens, suggesting cross-clade priming of NAb responses. In addition, vaccines encoding Env immunogens heterologous to SHIV-89.6P primed for a rapid anamnestic NAb response following infection compared to vaccines lacking an Env component. These results show that DNA/rAd immunization with multiple diverse Env immunogens is a viable approach for enhancing the breadth of NAb responses against HIV-1, and suggest that Env immunogens can prime for anamnestic NAb responses against a heterologous challenge virus.
Collapse
Affiliation(s)
- Michael S Seaman
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, 330 Brookline Ave/RE-204, Boston, MA 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Duke CM, Maguire CA, Keefer MC, Federoff HJ, Bowers WJ, Dewhurst S. HSV-1 amplicon vectors elicit polyfunctional T cell responses to HIV-1 Env, and strongly boost responses to an adenovirus prime. Vaccine 2007; 25:7410-21. [PMID: 17868958 PMCID: PMC2092414 DOI: 10.1016/j.vaccine.2007.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2007] [Revised: 08/03/2007] [Accepted: 08/07/2007] [Indexed: 12/11/2022]
Abstract
HSV-1 amplicon vectors elicit strong T-cell responses to encoded antigens but the qualitative nature of these responses is poorly understood. Antigen-specific CD4(+) and CD8(+) T-cell responses to amplicon and adenovirus (rAd5) vectors encoding HIV-1 gp120 were assessed following immunization of mice, by performing intracellular cytokine staining for IFNgamma, IL2 and TNFalpha, following stimulation of splenocytes with a HIV-1 Env peptide pool. The quality of the primary T-cell response to amplicon and rAd5 vectors was strikingly similar, but there were qualitative differences in responses to amplicon vectors that incorporated different promoters upstream of gp120 - suggesting that promoters can significantly influence immune response quality. When prime-boost combinations were studied, a rAd5 prime and amplicon boost elicited the highest T-cell response. Furthermore, protocols that incorporated a rAd5 prime consistently elicited a greater proportion of polyfunctional CD4(+) T-cells-regardless of boost. This suggests that initial priming can shape immune response quality after a boost. Overall, these findings provide insight into effective vector combinations for HIV-1 vaccine development.
Collapse
Affiliation(s)
- Cindy M.P. Duke
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Casey A. Maguire
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Michael C. Keefer
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Howard J. Federoff
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
- Department of Center for Aging and Development, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - William J. Bowers
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
- Department of Center for Aging and Development, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Stephen Dewhurst
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| |
Collapse
|
24
|
Santra S, Sun Y, Parvani JG, Philippon V, Wyand MS, Manson K, Gomez-Yafal A, Mazzara G, Panicali D, Markham PD, Montefiori DC, Letvin NL. Heterologous prime/boost immunization of rhesus monkeys by using diverse poxvirus vectors. J Virol 2007; 81:8563-70. [PMID: 17553898 PMCID: PMC1951337 DOI: 10.1128/jvi.00744-07] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
As the diversity of potential immunogens increases within certain classes of vectors, the possibility has arisen of employing heterologous prime/boost immunizations using diverse members of the same family of vectors. The present study was initiated to explore the use of divergent pox vectors in a prime/boost regimen to elicit high-frequency cellular immune responses to human immunodeficiency virus type 1 envelope and simian immunodeficiency virus gag in rhesus monkeys. We demonstrated that monkeys vaccinated with a recombinant modified vaccinia virus Ankara (rMVA) prime/recombinant fowlpox virus (rFPV) boost regimen and monkeys vaccinated with a recombinant vaccinia virus prime/rFPV boost regimen developed comparable cellular immune responses that were greater in magnitude than those elicited by a homologous prime/boost with rMVA. Nevertheless, comparable magnitude recall cellular immune responses were observed in monkeys vaccinated with heterologous and homologous recombinant poxvirus following challenge with the CXCR4-tropic SHIV-89.6P. Consistent with this finding, comparable levels of containment of viral replication and CD4(+) T-lymphocyte preservation were seen in these groups of recombinant poxvirus-vaccinated monkeys. This study supports further exploration of combining recombinant vectors of the same family in prime/boost immunization strategies to optimize vaccine-elicited cellular immune responses.
Collapse
Affiliation(s)
- Sampa Santra
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cristillo AD, Lisziewicz J, He L, Lori F, Galmin L, Trocio JN, Unangst T, Whitman L, Hudacik L, Bakare N, Whitney S, Restrepo S, Suschak J, Ferrari MG, Chung HK, Kalyanaraman VS, Markham P, Pal R. HIV-1 prophylactic vaccine comprised of topical DermaVir prime and protein boost elicits cellular immune responses and controls pathogenic R5 SHIV162P3. Virology 2007; 366:197-211. [PMID: 17499328 DOI: 10.1016/j.virol.2007.04.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 03/29/2007] [Accepted: 04/11/2007] [Indexed: 11/30/2022]
Abstract
Topical DNA vaccination (DermaVir) facilitates antigen presentation to naive T cells. DermaVir immunization in mice, using HIV-1 Env and Gag, elicited cellular immune responses. Boosting with HIV-1 gp120 Env and p41 Gag augmented Th1 cytokine levels. Intramuscular DNA administration was less efficient in priming antigen-specific cytokine production and memory T cells. In rhesus macaques, DermaVir immunization induced Gag- and Env-specific Th1 and Th2 cytokines and generation of memory T cells. Boosting of DermaVir-primed serum antibody levels was noted following gp140(SHIV89.6P)/p27(SIV) immunization. Rectal challenge with pathogenic R5-tropic SHIV162P3 resulted in control of plasma viremia (4/5 animals) that was reflected in jejunum, colon and mesenteric lymph nodes. An inverse correlation was found between Gag- and Env-specific central memory T cell responses on the day of challenge and plasma viremia at set point. Overall, the topical DermaVir/protein vaccination yields central memory T cell responses and facilitates control of pathogenic SHIV infection.
Collapse
Affiliation(s)
- Anthony D Cristillo
- Advanced BioScience Laboratories, Inc., 5510 Nicholson Lane, Kensington, MD 20895, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Thirugnanam S, Pandiaraja P, Ramaswamy K, Murugan V, Gnanasekar M, Nandakumar K, Reddy MVR, Kaliraj P. Brugia malayi: comparison of protective immune responses induced by Bm-alt-2 DNA, recombinant Bm-ALT-2 protein and prime-boost vaccine regimens in a jird model. Exp Parasitol 2007; 116:483-91. [PMID: 17442307 PMCID: PMC2763209 DOI: 10.1016/j.exppara.2007.02.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2006] [Revised: 02/28/2007] [Accepted: 02/28/2007] [Indexed: 11/30/2022]
Abstract
Immunization of jirds with Bm-alt-2 elicited partial protection against challenge infection with the filarial parasite Brugia malayi. In this study, we initially compared the protective immune responses elicited following immunization with recombinant Bm-ALT-2 protein regimen and Bm-alt-2 DNA regimen. These studies showed that protein vaccination conferred approximately 75% protection compared to DNA vaccination that conferred only 57% protection. Analysis of the protective immune responses showed that the protein immunization promoted a Th2-biased response with an increase in IL-4, IL-5 and IgG1 responses, whereas, the DNA vaccine promoted a Th1-biased response with profound IFN-gamma and IgG2a responses. Since protein vaccination gave better results than DNA vaccination, we then wanted to evaluate whether a prime-boost vaccination that combined DNA prime and protein boost will significantly increase the protective responses induced by the protein vaccine. Our results suggest that prime-boost vaccination had no added advantage and was comparatively less effective (64% protection) than the Bm-ALT-2 protein alone vaccination. Prime boost vaccination generated mixed Th1/Th2 responses with a slightly diminished Th2 responses compared to protein vaccination. Thus, our results suggest that Bm-ALT-2 protein vaccination regimen may be slightly better than prime-boost vaccine regimen and the mechanism of protection appears to be largely mediated by a Th2-biased response.
Collapse
MESH Headings
- Animals
- Antibodies, Helminth/biosynthesis
- Antibodies, Helminth/blood
- Antigens, Helminth/genetics
- Antigens, Helminth/immunology
- Brugia malayi/immunology
- Cytokines/biosynthesis
- Cytokines/genetics
- DNA, Helminth/immunology
- Diffusion Chambers, Culture
- Disease Models, Animal
- Elephantiasis, Filarial/prevention & control
- Gerbillinae
- Helminth Proteins/genetics
- Helminth Proteins/immunology
- Immunity, Cellular
- Immunization/methods
- Lymphocyte Activation
- Male
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Th2 Cells/immunology
- Vaccines/immunology
- Vaccines, DNA/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
| | | | - Kalyanasundaram Ramaswamy
- Department of Biomedical Sciences, University of Illinois, College of Medicine, Rockford, IL 61107, USA
| | - Vadivel Murugan
- Centre for Biotechnology, Anna University, Chennai 600 025, India
| | - Munirathinam Gnanasekar
- Department of Biomedical Sciences, University of Illinois, College of Medicine, Rockford, IL 61107, USA
| | - Krithika Nandakumar
- Jamnalal Bajaj Tropical Disease Research Centre, Mahatma Gandhi Institute of Medical Sciences, Sevagram, India
| | - Maryada Venkata Rami Reddy
- Jamnalal Bajaj Tropical Disease Research Centre, Mahatma Gandhi Institute of Medical Sciences, Sevagram, India
- Corresponding authors. Fax: +91 44 22542299. (M.V.R. Reddy), (P. Kaliraj)
| | - Perumal Kaliraj
- Centre for Biotechnology, Anna University, Chennai 600 025, India
- Corresponding authors. Fax: +91 44 22542299. (M.V.R. Reddy), (P. Kaliraj)
| |
Collapse
|
27
|
Hanke T, McMichael AJ, Dorrell L. Clinical experience with plasmid DNA- and modified vaccinia virus Ankara-vectored human immunodeficiency virus type 1 clade A vaccine focusing on T-cell induction. J Gen Virol 2007; 88:1-12. [PMID: 17170430 DOI: 10.1099/vir.0.82493-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Candidate human immunodeficiency virus type 1 (HIV-1) vaccines focusing on T-cell induction, constructed as pTHr.HIVA DNA and modified vaccinia virus Ankara (MVA).HIVA, were delivered in a heterologous prime-boost regimen. The vaccines were tested in several hundred healthy or HIV-1-infected volunteers in Europe and Africa. Whilst larger trials of hundreds of volunteers suggested induction of HIV-1-specific T-cell responses in <15 % of healthy vaccinees, a series of small, rapid trials in 12-24 volunteers at a time with a more in-depth analysis of vaccine-elicited T-cell responses proved to be highly informative and provided more encouraging results. These trials demonstrated that the pTHr.HIVA vaccine alone primed consistently weak and mainly CD4(+), but also CD8(+) T-cell responses, and the MVA.HIVA vaccine delivered a consistent boost to both CD4(+) and CD8(+) T cells, which was particularly strong in HIV-1-infected patients. Thus, whilst the search is on for ways to enhance T-cell priming, MVA is a useful boosting vector for human subunit genetic vaccines.
Collapse
Affiliation(s)
- Tomáš Hanke
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| | - Andrew J McMichael
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| | - Lucy Dorrell
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| |
Collapse
|
28
|
Demberg T, Florese RH, Heath MJ, Larsen K, Kalisz I, Kalyanaraman VS, Lee EM, Pal R, Venzon D, Grant R, Patterson LJ, Korioth-Schmitz B, Buzby A, Dombagoda D, Montefiori DC, Letvin NL, Cafaro A, Ensoli B, Robert-Guroff M. A replication-competent adenovirus-human immunodeficiency virus (Ad-HIV) tat and Ad-HIV env priming/Tat and envelope protein boosting regimen elicits enhanced protective efficacy against simian/human immunodeficiency virus SHIV89.6P challenge in rhesus macaques. J Virol 2007; 81:3414-27. [PMID: 17229693 PMCID: PMC1866031 DOI: 10.1128/jvi.02453-06] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We previously demonstrated that replication-competent adenovirus (Ad)-simian immunodeficiency virus (SIV) recombinant prime/protein boost regimens elicit potent immunogenicity and strong, durable protection of rhesus macaques against SIV(mac251). Additionally, native Tat vaccines have conferred strong protection against simian/human immunodeficiency virus SHIV(89.6P) challenge of cynomolgus monkeys, while native, inactivated, or vectored Tat vaccines have failed to elicit similar protective efficacy in rhesus macaques. Here we asked if priming rhesus macaques with replicating Ad-human immunodeficiency virus (HIV) tat and boosting with the Tat protein would elicit protection against SHIV(89.6P). We also evaluated a Tat/Env regimen, adding an Ad-HIV env recombinant and envelope protein boost to test whether envelope antibodies would augment acute-phase protection. Further, expecting cellular immunity to enhance chronic viremia control, we tested a multigenic group: Ad-HIV tat, -HIV env, -SIV gag, and -SIV nef recombinants and Tat, Env, and Nef proteins. All regimens were immunogenic. A hierarchy was observed in enzyme-linked immunospot responses (with the strongest response for Env, followed by Gag, followed by Nef, followed by Tat) and antibody titers (with the highest titer for Env, followed by Tat, followed by Nef, followed by Gag). Following intravenous SHIV(89.6P) challenge, all macaques became infected. Compared to controls, no protection was seen in the Tat-only group, confirming previous reports for rhesus macaques. However, the multigenic group blunted acute viremia by approximately 1 log (P = 0.017), and both the multigenic and Tat/Env groups reduced chronic viremia by 3 and 4 logs, respectively, compared to controls (multigenic, P = 0.0003; Tat/Env, P < 0.0001). The strikingly greater reduction in the Tat/Env group than in the multigenic group (P = 0.014) was correlated with Tat and Env binding antibodies. Since prechallenge anti-Env antibodies lacked SHIV(89.6P)-neutralizing activity, other functional anti-Env and anti-Tat activities are under investigation, as is a possible synergy between the Tat and Env immunogens.
Collapse
Affiliation(s)
- Thorsten Demberg
- Vaccine Branch, National Cancer Institute/NIH, 41 Medlars Drive, Building 41, Bethesda, MD 20892-5065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Manuel ER, Charini WA, Sen P, Peyerl FW, Kuroda MJ, Schmitz JE, Autissier P, Sheeter DA, Torbett BE, Letvin NL. Contribution of T-cell receptor repertoire breadth to the dominance of epitope-specific CD8+ T-lymphocyte responses. J Virol 2006; 80:12032-40. [PMID: 17035327 PMCID: PMC1676269 DOI: 10.1128/jvi.01479-06] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dominant epitope-specific CD8(+) T-lymphocyte responses play a central role in controlling viral spread. We explored the basis for the development of this focused immune response in simian immunodeficiency virus (SIV)- and simian-human immunodeficiency virus (SHIV)-infected rhesus monkeys through the use of two dominant (p11C and p199RY) and two subdominant (p68A and p56A) epitopes. Using real-time PCR to quantitate T-cell receptor (TCR) variable region beta (Vbeta) family usage, we show that CD8(+) T-lymphocyte populations specific for dominant epitopes are characterized by a diverse Vbeta repertoire, whereas those specific for subdominant epitopes employ a dramatically more focused Vbeta repertoire. We also demonstrate that dominant epitope-specific CD8(+) T lymphocytes employ TCRs with multiple CDR3 lengths, whereas subdominant epitope-specific cells employ TCRs with a more restricted CDR3 length. Thus, the relative dominance of an epitope-specific CD8(+) T-lymphocyte response reflects the clonal diversity of that response. These findings suggest that the limited clonal repertoire of subdominant epitope-specific CD8(+) T-lymphocyte populations may limit the ability of these epitope-specific T-lymphocyte populations to expand and therefore limit the ability of these cell populations to contribute to the control of viral replication.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Chromatography, High Pressure Liquid
- DNA Primers
- DNA, Complementary/genetics
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Flow Cytometry
- Genes, T-Cell Receptor beta/genetics
- Immunity, Cellular/immunology
- Macaca mulatta
- Molecular Sequence Data
- Peptides/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Sequence Analysis, DNA
- Simian Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Edwin R Manuel
- Beth Israel Deaconess Medical Center, 41 Ave. Louis Pasteur, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Im EJ, Nkolola JP, di Gleria K, McMichael AJ, Hanke T. Induction of long-lasting multi-specific CD8+T cells by a four-component DNA-MVA/HIVA-RENTA candidate HIV-1 vaccine in rhesus macaques. Eur J Immunol 2006; 36:2574-84. [PMID: 17013988 DOI: 10.1002/eji.200636482] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As a part of a long-term effort to develop vaccine against HIV-1 clade A inducing protective T cell responses in humans, we run mutually complementing studies in humans and non-human primates (NHP) with the aim to maximize vaccine immunogenicity. The candidate vaccine under development has four components, pTHr.HIVA and pTH.RENTA DNA, and modified vaccinia virus Ankara (MVA).HIVA and MVA.RENTA, delivered in a heterologous DNA prime-MVA boost regimen. While the HIVA (Gag/epitopes) components have been tested in NHP and over 300 human subjects, we plan to test in humans the RENTA (reverse transcriptase, gp41, Nef, Tat) vaccines designed to broaden HIVA-induced responses in year 2007. Here, we investigated the four-component vaccine long-term immunogenicity in Mamu-A*01-positive rhesus macaques and demonstrated that the vaccine-induced T cells were multi-specific, multi-functional, readily proliferated to recall peptides and were circulating in the peripheral blood of vaccine recipients over 1 year after vaccine administration. The consensus clade A-elicited T cells recognized 50% of tested epitope variants from other HIV-1 clades. Thus, the DNA-MVA/HIVA-RENTA vaccine induced memory T cells of desirable characteristics and similarities to those induced in humans by HIVA vaccines alone; however, single-clade vaccines may not elicit sufficiently cross-reactive responses.
Collapse
Affiliation(s)
- Eung-Jun Im
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
31
|
Luzuriaga K, Newell ML, Dabis F, Excler JL, Sullivan JL. Vaccines to prevent transmission of HIV-1 via breastmilk: scientific and logistical priorities. Lancet 2006; 368:511-21. [PMID: 16890838 DOI: 10.1016/s0140-6736(06)69159-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mother-to-child transmission (MTCT) of HIV-1 is the major mode of paediatric infection. The rapidly increasing incidence of MTCT worldwide has resulted in an urgent need for preventive strategies. Antiretroviral regimens can prevent intrapartum HIV transmission; however, these regimens do not prevent HIV transmission through breastfeeding. Furthermore, children who escape MTCT are again at risk of infection when they become sexually active as adolescents. An infant vaccine regimen, begun at birth, would hence be a more attractive strategy and might also provide the basis for lifetime protection. Unique features of MTCT and paediatric HIV disease could be helpful in understanding correlates of immune protection and could facilitate rapid assessment of vaccine efficacy. Thus, there is compelling rationale to develop safe, effective HIV vaccines for use in infants and children. Here, we discuss the scientific and logistical challenges for the development of paediatric HIV vaccines; available vaccines and completed or planned paediatric vaccine trials are also discussed.
Collapse
Affiliation(s)
- Katherine Luzuriaga
- University of Massachusetts Medical School, Program in Molecular Medicine, 373 Plantation Street, Suite 318, Worcester, MA 01605, USA.
| | | | | | | | | |
Collapse
|
32
|
Dorrell L, Yang H, Ondondo B, Dong T, di Gleria K, Suttill A, Conlon C, Brown D, Williams P, Bowness P, Goonetilleke N, Rostron T, Rowland-Jones S, Hanke T, McMichael A. Expansion and diversification of virus-specific T cells following immunization of human immunodeficiency virus type 1 (HIV-1)-infected individuals with a recombinant modified vaccinia virus Ankara/HIV-1 Gag vaccine. J Virol 2006; 80:4705-16. [PMID: 16641264 PMCID: PMC1472080 DOI: 10.1128/jvi.80.10.4705-4716.2006] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Affordable therapeutic strategies that induce sustained control of human immunodeficiency virus type 1 (HIV-1) replication and are tailored to the developing world are urgently needed. Since CD8(+) and CD4(+) T cells are crucial to HIV-1 control, stimulation of potent cellular responses by therapeutic vaccination might be exploited to reduce antiretroviral drug exposure. However, therapeutic vaccines tested to date have shown modest immunogenicity. In this study, we performed a comprehensive analysis of the changes in virus-specific CD8(+) and CD4(+) T-cell responses occurring after vaccination of 16 HIV-1-infected individuals with a recombinant modified vaccinia virus Ankara-vectored vaccine expressing the consensus HIV-1 clade A Gag p24/p17 sequences and multiple CD8(+) T-cell epitopes during highly active antiretroviral therapy. We observed significant amplification and broadening of CD8(+) and CD4(+) gamma interferon responses to vaccine-derived epitopes in the vaccinees, without rebound viremia, but not in two unvaccinated controls followed simultaneously. Vaccine-driven CD8(+) T-cell expansions were also detected by tetramer reactivity, predominantly in the CD45RA(-) CCR7(+) or CD45RA(-) CCR7(-) compartments, and persisted for at least 1 year. Expansion was associated with a marked but transient up-regulation of CD38 and perforin within days of vaccination. Gag-specific CD8(+) and CD4(+) T-cell proliferation also increased postvaccination. These data suggest that immunization with MVA.HIVA is a feasible strategy to enhance potentially protective T-cell responses in individuals with chronic HIV-1 infection.
Collapse
Affiliation(s)
- Lucy Dorrell
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Brander C, Frahm N, Walker BD. The challenges of host and viral diversity in HIV vaccine design. Curr Opin Immunol 2006; 18:430-7. [PMID: 16777397 DOI: 10.1016/j.coi.2006.05.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2006] [Accepted: 05/31/2006] [Indexed: 02/03/2023]
Abstract
Rational HIV vaccine design is crucially dependent on a number of factors, including a detailed understanding of the immune responses that control infection in individuals that have non-progressing disease, the impact of host genetics on these responses, and the degree of immunological cross-reactivity between the vaccine immunogen and the encountered virus antigens. Significant progress has been made in a number of these areas over the past five years, which might help in the generation of a more effective immunogen design and will provide opportunities for novel vaccine delivery options. However, the understanding of immune response(s) that can mediate protection from infection or, if infection ensues, that slow the rate of HIV disease progression is still incomplete and will require detailed studies in unprecedentedly large populations infected with different HIV clades, combining advances in virology, immunology, human host genetics and bioinformatics analyses for the optimal design of vaccine immunogens.
Collapse
Affiliation(s)
- Christian Brander
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School Charlestown, 02192, USA.
| | | | | |
Collapse
|
34
|
Acierno PM, Schmitz JE, Gorgone DA, Sun Y, Santra S, Seaman MS, Newberg MH, Mascola JR, Nabel GJ, Panicali D, Letvin NL. Preservation of functional virus-specific memory CD8+ T lymphocytes in vaccinated, simian human immunodeficiency virus-infected rhesus monkeys. THE JOURNAL OF IMMUNOLOGY 2006; 176:5338-45. [PMID: 16622001 DOI: 10.4049/jimmunol.176.9.5338] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Functional impairment of virus-specific memory CD8(+) T lymphocytes has been associated with clinical disease progression following HIV, SIV, and simian human immunodeficiency virus infection. These lymphocytes have a reduced capacity to produce antiviral cytokines and mediators involved in the lysis of virally infected cells. In the present study, we used polychromatic flow cytometry to assess the frequency and functional capacity of central memory (CD28(+)CD95(+)) and effector memory (CD28(-)CD95(+)) subpopulations of Gag-specific CD8(+) T cells in SIV/simian human immunodeficiency virus-infected rhesus monkeys. The aim of this study was to determine whether Ag-specific, memory CD8(+) T cell function could be preserved in infected monkeys that had been immunized before infection with a vaccine regimen consisting of a plasmid DNA prime followed by a recombinant viral vector boost. We observed that vaccination was associated with the preservation of Gag-specific central memory CD8(+) T cells that were functionally capable of producing IFN-gamma, and effector memory CD8(+) T cells that were capable of producing granzyme B following viral Ag exposure.
Collapse
Affiliation(s)
- Paula M Acierno
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Elbasha EH, Gumel AB. Theoretical Assessment of Public Health Impact of Imperfect Prophylactic HIV-1 Vaccines with Therapeutic Benefits. Bull Math Biol 2006; 68:577-614. [PMID: 16794946 DOI: 10.1007/s11538-005-9057-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Accepted: 04/20/2005] [Indexed: 10/24/2022]
Abstract
This paper presents a number of deterministic models for theoretically assessing the potential impact of an imperfect prophylactic HIV-1 vaccine that has five biological modes of action, namely "take," "degree," "duration," "infectiousness," and "progression," and can lead to increased risky behavior. The models, which are of the form of systems of nonlinear differential equations, are constructed via a progressive refinement of a basic model to incorporate more realistic features of HIV pathogenesis and epidemiology such as staged progression, differential infectivity, and HIV transmission by AIDS patients. The models are analyzed to gain insights into the qualitative features of the associated equilibria. This allows the determination of important epidemiological thresholds such as the basic reproduction numbers and a measure for vaccine impact or efficacy. The key findings of the study include the following (i) if the vaccinated reproduction number is greater than unity, each of the models considered has a locally unstable disease-free equilibrium and a unique endemic equilibrium; (ii) owing to the vaccine-induced backward bifurcation in these models, the classical epidemiological requirement of vaccinated reproduction number being less than unity does not guarantee disease elimination in these models; (iii) an imperfect vaccine will reduce HIV prevalence and mortality if the reproduction number for a wholly vaccinated population is less than the corresponding reproduction number in the absence of vaccination; (iv) the expressions for the vaccine characteristics of the refined models take the same general structure as those of the basic model.
Collapse
Affiliation(s)
- Elamin H Elbasha
- Merck Research Laboratories, 10 Sentry Parkway, BL 2-3, Blue Bell, PA 19422, USA.
| | | |
Collapse
|
36
|
Abaitua F, Rodríguez JR, Garzón A, Rodríguez D, Esteban M. Improving recombinant MVA immune responses: Potentiation of the immune responses to HIV-1 with MVA and DNA vectors expressing Env and the cytokines IL-12 and IFN-gamma. Virus Res 2006; 116:11-20. [PMID: 16214252 DOI: 10.1016/j.virusres.2005.08.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Revised: 07/22/2005] [Accepted: 08/22/2005] [Indexed: 11/23/2022]
Abstract
Recombinants based on vaccinia virus vectors, especially on the highly attenuated modified vaccinia virus Ankara (MVA) strain, are now being tested in clinical trials for safety and immunogenicity, using prime/boost heterologous regimes of vaccination. Due to the limited replication capacity of MVA, it is necessary to develop procedures that can enhance the specific cellular immune responses to the recombinant antigen delivered by the MVA vector. In this investigation, we have characterized the systemic immune responses in BALB/c mice using interferon-gamma (IFN-gamma) or interleukin-12 (IL-12) in an adjuvant-like manner elicited by MVA recombinants or naked DNA vectors expressing one of those cytokines in combination with the human immunodeficiency virus type 1 (HIV-1) envelope (Env) as antigen. In infected mice, virus gene expression in splenocytes and levels of cytokines IFN-gamma and IL-12 in serum were maximal by 6h post-infection (hpi) with MVA recombinants expressing IFN-gamma (MVAIFN-gamma) or IL-12 (MVAIL-12). In the infected animals, co-expression of HIV-1 env (MVAENV) and either IFN-gamma or IL-12 from MVA recombinants produced a two and three-fold increase of anti-env CD8+ T cell response, respectively. When priming was carried out with DNA vectors expressing HIV-1 env and either IFN-gamma or IL-12, the magnitude of the specific anti-env CD8+ T cell stimulation after MVAENV booster was further enhanced. Our findings revealed that IFN-gamma or IL-12 can be used to potentiate the cellular immune response to HIV-1 env, when delivered either from a single MVA recombinant or from a DNA vector. The increment of the CD8+ T cell response was higher in a DNA/MVA prime/boost protocol. Thus, the immune response of MVA vectors can be improved with the co-delivery of the cytokines IFN-gamma or IL-12.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Adjuvants, Immunologic/genetics
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cytokines/biosynthesis
- DNA, Viral
- Female
- Gene Products, env/genetics
- Gene Products, env/immunology
- Genetic Vectors/genetics
- Genetic Vectors/immunology
- HIV-1/immunology
- Immunization, Secondary
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interleukin-12/genetics
- Interleukin-12/immunology
- Mice
- Mice, Inbred BALB C
- Models, Animal
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Fernando Abaitua
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Campus Ciudad Universitaria Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | |
Collapse
|
37
|
Abstract
HIV poses a serious health threat in the world. Mucosal transmission of HIV through the genitourinary tract may be the most important route of transmission. Intranasal immunisations induce vaginal and systemic immune responses. Various protein-, DNA- and RNA-based immunopotentiating adjuvants/delivery systems and live bacterial and viral vectors are available for intranasal immunisations, and these systems may differ in their ability to induce a specific type of immune response (e.g., a cytotoxic T cell versus an antibody response). As the protection against HIV may require both cytotoxic T cell and antibodies, a combination of adjuvants/delivery systems for combinations of mucosal and parenteral immunisations may be required in order to develop a protective anti-HIV vaccine.
Collapse
Affiliation(s)
- Michael Vajdy
- Chiron Vaccines, 4560 Horton Street, Emeryville, CA 94608, USA.
| | | |
Collapse
|
38
|
Bryant KJ. Expanding research on the role of alcohol consumption and related risks in the prevention and treatment of HIV/AIDS. Subst Use Misuse 2006; 41:1465-507. [PMID: 17002990 DOI: 10.1080/10826080600846250] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This article is a review of some of the major epidemiological, behavioral, biological, and integrative prevention research issues and priorities in the area of HIV/AIDS and alcohol consumption. Drinking alcohol increases both the risk for infection with HIV and related illnesses and the morbidity and mortality of patients who progress to AIDS. New and improved measurement procedures have helped in assessment of the complex patterns of alcohol use, identification of intervening explanatory mechanisms for risk behaviors and contexts, and determination of intervention outcomes. Both the direct and indirect effects of alcohol misuse appear to be major contributors to both the risk for infection with HIV and the transmission of HIV/AIDS at the individual and population levels. There is increasing evidence that perhaps no level of alcohol consumption is "safe" for those who are HIV infected and receiving antiretroviral treatment. Interdisciplinary basic behavioral and biomedical research is needed to develop comprehensive culturally appropriate strategies for programs that can be effectively delivered in community contexts in the United States and abroad and that focus on the integration of our understanding of individual behaviors, high-risk group membership, biological mechanisms, and the social and physical environments that place individuals at risk for HIV infection. High-priority topics include improving adherence to antiretroviral medications, prevention of infection in young minority women in the United States, and treatment of HIV+ pregnant women who are alcohol abusers to prevent adverse fetal outcomes, which is an international focus in under-resourced settings in Africa.
Collapse
|
39
|
Cristillo AD, Wang S, Caskey MS, Unangst T, Hocker L, He L, Hudacik L, Whitney S, Keen T, Chou THW, Shen S, Joshi S, Kalyanaraman VS, Nair B, Markham P, Lu S, Pal R. Preclinical evaluation of cellular immune responses elicited by a polyvalent DNA prime/protein boost HIV-1 vaccine. Virology 2005; 346:151-68. [PMID: 16325880 DOI: 10.1016/j.virol.2005.10.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 09/19/2005] [Accepted: 10/21/2005] [Indexed: 10/25/2022]
Abstract
While DNA vaccines have been shown to prime cellular immune responses, levels are often low in nonhuman primates or humans. Hence, efforts have been directed toward boosting responses by combining DNA with different vaccination modalities. To this end, a polyvalent DNA prime/protein boost vaccine, consisting of codon optimized HIV-1 env (A, B, C, E) and gag (C) and homologous gp120 proteins in QS-21, was evaluated in rhesus macaques and BALB/c mice. Humoral and cellular responses, detected following DNA immunization, were increased following protein boost in macaques and mice. In dissecting cellular immune responses in mice, protein-enhanced responses were found to be mediated by CD4+ and CD8+ T cells with a Th1 cytokine bias. Our study reveals that, in addition to augmenting humoral responses, protein boosting of DNA-primed animals augments cellular immune responses mediated by CD8+ CTL, CD4+ T-helper cells and Th1 cytokines; thus, offering much promise in controlling HIV-1 in vaccinees.
Collapse
Affiliation(s)
- Anthony D Cristillo
- Advanced BioScience Laboratories, Department of Cell Biology, 5510 Nicholson Lane, Kensington, MD 20895, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Keating SM, Bejon P, Berthoud T, Vuola JM, Todryk S, Webster DP, Dunachie SJ, Moorthy VS, McConkey SJ, Gilbert SC, Hill AVS. Durable human memory T cells quantifiable by cultured enzyme-linked immunospot assays are induced by heterologous prime boost immunization and correlate with protection against malaria. THE JOURNAL OF IMMUNOLOGY 2005; 175:5675-80. [PMID: 16237057 DOI: 10.4049/jimmunol.175.9.5675] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Immunological memory is a required component of protective antimalarial responses raised by T cell-inducing vaccines. The magnitude of ex vivo IFN-gamma T cell responses is widely used to identify immunogenic vaccines although this response usually wanes and may disappear within weeks. However, protection in the field is likely to depend on durable central memory T cells that are not detected by this assay. To identify longer-lived memory T cells, PBMC from malaria-naive vaccinated volunteers who had received prime boost vaccinations with a combination of DNA and/or viral vectors encoding the multiepitope string-thrombospondin-related adhesion protein Ag were cultured in vitro with Ag for 10 days before the ELISPOT assay. Ex vivo T cell responses peaked at 7 days after the final immunization and declined substantially over 6 mo, but responses identified after T cell culture increased over the 6-mo period after the final immunization. Moreover, individual cultured ELISPOT responses at the day of challenge time point correlated significantly with degree of protection against malaria sporozoite challenge, whereas ex vivo responses did not, despite a correlation between the peak ex vivo response and magnitude of memory responses 6 mo later. This cultured assay identifies long-lasting protective T cell responses and therefore offers an attractive option for assessments of vaccine immunogenicity.
Collapse
Affiliation(s)
- Sheila M Keating
- Centre for Clinical Vaccinology and Tropical Medicine, Nuffield Department of Medicine, Churchill Hospital, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Johnston RE, Johnson PR, Connell MJ, Montefiori DC, West A, Collier ML, Cecil C, Swanstrom R, Frelinger JA, Davis NL. Vaccination of macaques with SIV immunogens delivered by Venezuelan equine encephalitis virus replicon particle vectors followed by a mucosal challenge with SIVsmE660. Vaccine 2005; 23:4969-79. [PMID: 16005121 DOI: 10.1016/j.vaccine.2005.05.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Revised: 05/25/2005] [Accepted: 05/27/2005] [Indexed: 11/18/2022]
Abstract
VEE replicon particles (VRP), non-propagating vaccine vectors derived from Venezuelan equine encephalitis virus (VEE), were engineered to express immunogens from the cloned isolate SIVsmH-4, combined in a vaccine cocktail and inoculated subcutaneously to immunize rhesus macaques. The virulent, uncloned challenge stock, SIVsmE660, represented a type of heterologous challenge and the intrarectal challenge modeled infection across a mucosal surface. Prechallenge neutralizing antibodies against SIVsmH-4 were induced in all vaccinates, and a prechallenge cellular immune response could be detected in one of six. Post-challenge, virus loads were reduced at the peak, at set point and at termination (41 weeks post-challenge), although these differences did not reach statistical significance. Significantly elevated levels of CD4+ T cells were observed post-challenge. A strong correlation was noted between a net increase in CD4+ T cell count and lowered virus load at set point.
Collapse
Affiliation(s)
- Robert E Johnston
- Carolina Vaccine Institute, School of Medicine, University of North Carolina, CB#7292, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wu L, Kong WP, Nabel GJ. Enhanced breadth of CD4 T-cell immunity by DNA prime and adenovirus boost immunization to human immunodeficiency virus Env and Gag immunogens. J Virol 2005; 79:8024-31. [PMID: 15956548 PMCID: PMC1143709 DOI: 10.1128/jvi.79.13.8024-8031.2005] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A variety of gene-based vaccination approaches have been used to enhance the immune response to viral pathogens. Among them, the ability to perform heterologous immunization by priming with DNA and boosting with replication-defective adenoviral (ADV) vectors encoding foreign antigens has proven particularly effective in eliciting enhanced cellular and humoral immunity compared to either agent alone. Because adenoviral vector immunization alone can elicit substantial cellular and humoral immune responses in a shorter period of time, we asked whether the immune response induced by the prime-boost immunization was different from adenoviral vaccines with respect to the potency and breadth of T-cell recognition. While DNA/ADV immunization stimulated the CD8 response, it was directed to the same epitopes in Gag and Env immunogens of human immunodeficiency virus as DNA or ADV alone. In contrast, the CD4 response to these immunogens diversified after DNA/ADV immunization compared to each vector alone. These findings suggest that the diversity of the CD4 immune response is increased by DNA/ADV prime-boost vaccination and that these components work synergistically to enhance T-cell epitope recognition.
Collapse
Affiliation(s)
- Lan Wu
- Vaccine Research Center, NIAID, National Institutes of Health, Room 4502, Bldg. 40, MSC-3005, 40 Convent Drive, Bethesda, MD 20892-3005, USA
| | | | | |
Collapse
|
43
|
Abstract
Vaccination, or the deliberate induction of protective immunity by administering nonpathogenic forms of a microbe or its antigens to induce a memory immune response, is the world's most cost-effective medical procedure for preventing morbidity and mortality caused by infectious disease. Historically, most vaccines have worked by eliciting long-lived plasma cells. These cells produce antibodies that limit disease by neutralizing a toxin or blocking the spread of the infectious agent. For these 'B cell vaccines,' the immunological marker, or correlate, for protection is the titer of protective antibodies. With the discovery of HIV/AIDS, vaccine development has been confronted by an agent that is not easily blocked by antibody. To overcome this, researchers who are developing HIV/AIDS vaccines have turned to the elicitation of cellular immunity, or 'T cell vaccines,' which recognize and kill infected cells.
Collapse
Affiliation(s)
- Harriet L Robinson
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329, USA.
| | | |
Collapse
|
44
|
Jackson SS, Ilyinskii P, Philippon V, Gritz L, Yafal AG, Zinnack K, Beaudry KR, Manson KH, Lifton MA, Kuroda MJ, Letvin NL, Mazzara GP, Panicali DL. Role of genes that modulate host immune responses in the immunogenicity and pathogenicity of vaccinia virus. J Virol 2005; 79:6554-9. [PMID: 15858042 PMCID: PMC1091706 DOI: 10.1128/jvi.79.10.6554-6559.2005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Poxvirus vaccine vectors, although capable of eliciting potent immune responses, pose serious health risks in immunosuppressed individuals. We therefore constructed five novel recombinant vaccinia virus vectors which contained overlapping deletions of coding regions for the B5R, B8R, B12R, B13R, B14R, B16R, B18R, and B19R immunomodulatory gene products and assessed them for both immunogenicity and pathogenicity. All five of these novel vectors elicited both cellular and humoral immunity to the inserted HIV-BH10 env comparable to that induced by the parental Wyeth strain vaccinia virus. However, deletion of these immunomodulatory genes did not increase the immunogenicity of these vectors compared with the parental vaccinia virus. Furthermore, four of these vectors were slightly less virulent and one was slightly more virulent than the Wyeth strain virus in neonatal mice. Attenuated poxviruses have potential use as safer alternatives to current replication-competent vaccinia virus. Improved vaccinia virus vectors can be generated by deleting additional genes to achieve a more significant viral attenuation.
Collapse
Affiliation(s)
- Shawn S Jackson
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Research East -RE 113, 330 Brookline Ave., Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Santra S, Seaman MS, Xu L, Barouch DH, Lord CI, Lifton MA, Gorgone DA, Beaudry KR, Svehla K, Welcher B, Chakrabarti BK, Huang Y, Yang ZY, Mascola JR, Nabel GJ, Letvin NL. Replication-defective adenovirus serotype 5 vectors elicit durable cellular and humoral immune responses in nonhuman primates. J Virol 2005; 79:6516-22. [PMID: 15858035 PMCID: PMC1091731 DOI: 10.1128/jvi.79.10.6516-6522.2005] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The magnitude and durability of immune responses induced by replication-defective adenovirus serotype 5 (ADV5) vector-based vaccines were evaluated in the simian-human immunodeficiency virus/rhesus monkey model. A single inoculation of recombinant ADV5 vector constructs induced cellular and humoral immunity, but the rapid generation of neutralizing anti-Ad5 antibodies limited the immunity induced by repeated vector administration. The magnitude and durability of the immune responses elicited by these vaccines were greater when they were delivered as boosting immunogens in plasmid DNA-primed monkeys than when they were used as single-modality immunogens. Therefore, administration of ADV5-based vectors in DNA-primed subjects may be a preferred use of this vaccine modality for generating long-term immune protection.
Collapse
Affiliation(s)
- Sampa Santra
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, RE113, P. O. Box 15732, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Seaman MS, Santra S, Newberg MH, Philippon V, Manson K, Xu L, Gelman RS, Panicali D, Mascola JR, Nabel GJ, Letvin NL. Vaccine-elicited memory cytotoxic T lymphocytes contribute to Mamu-A*01-associated control of simian/human immunodeficiency virus 89.6P replication in rhesus monkeys. J Virol 2005; 79:4580-8. [PMID: 15795244 PMCID: PMC1069575 DOI: 10.1128/jvi.79.8.4580-4588.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The expression of particular major histocompatibility complex (MHC) class I alleles can influence the rate of disease progression following lentiviral infections. This effect is a presumed consequence of potent cytotoxic T-lymphocyte (CTL) responses that are restricted by these MHC class I molecules. The present studies have examined the impact of the MHC class I allele Mamu-A*01 on simian/human immunodeficiency virus 89.6P (SHIV-89.6P) infection in unvaccinated and vaccinated rhesus monkeys by exploring the contribution of dominant-epitope specific CTL in this setting. Expression of Mamu-A*01 in immunologically naive monkeys was not associated with improved control of viral replication, CD4+ T-lymphocyte loss, or survival. In contrast, Mamu-A*01+ monkeys that had received heterologous prime/boost immunizations prior to challenge maintained higher CD4+ T-lymphocyte levels and better control of SHIV-89.6P replication than Mamu-A*01- monkeys. This protection was associated with the evolution of high-frequency anamnestic CTL responses specific for a dominant Mamu-A*01-restricted Gag epitope following infection. These data indicate that specific MHC class I alleles can confer protection in the setting of a pathogenic SHIV infection by their ability to elicit memory CTL following vaccination.
Collapse
Affiliation(s)
- Michael S Seaman
- Beth Israel Deaconess Medical Center, Division of Viral Pathogenesis, 330 Brookline Ave. RE-113, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tscharke DC, Suhrbier A. From mice to humans – murine intelligence for human CD8+T cell vaccine design. Expert Opin Biol Ther 2005; 5:263-71. [PMID: 15757387 DOI: 10.1517/14712598.5.2.263] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There has been considerable progress in the design of vaccines capable of safely and effectively inducing CD8(+) T cells for prophylaxis and treatment of chronic infectious diseases and cancer. Much of what is known about CD8(+) T cell-mediated immunity has come from pioneering work in mice; this broad overview discusses recent work in mouse systems where lessons may be drawn for human vaccine development. The areas highlighted include antivector immunity, immunodominance, dendritic cell biology and targeting, the role of Toll-like receptors and their exploitation by novel adjuvants, the role of CD4(+) T cell help, regulatory T cells and, finally, some comments on the different requirements of prophylactic versus therapeutic vaccines.
Collapse
Affiliation(s)
- David C Tscharke
- Queensland Institute of Medical Research, EBVBiology, P.O. Royal Brisbane Hospital, Brisbane, QLD 4029, Australia.
| | | |
Collapse
|
48
|
De Rose R, Chea S, Dale CJ, Reece J, Fernandez CS, Wilson KM, Thomson S, Ramshaw IA, Coupar BEH, Boyle DB, Sullivan MT, Kent SJ. Subtype AE HIV-1 DNA and recombinant Fowlpoxvirus vaccines encoding five shared HIV-1 genes: safety and T cell immunogenicity in macaques. Vaccine 2005; 23:1949-56. [PMID: 15734067 DOI: 10.1016/j.vaccine.2004.10.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2004] [Revised: 10/08/2004] [Accepted: 10/14/2004] [Indexed: 11/28/2022]
Abstract
To induce broad T cell immunity to HIV-1, we evaluated the safety, immunogenicity and dose-response relationship of DNA and recombinant Fowlpoxvirus (rFPV) vaccines encoding five shared HIV subtype AE genes (Gag, Pol, Env, Tat, Rev) in pigtail macaques. The DNA (three doses of either 1 mg or 4.5 mg) and rFPV (a single boost of either 5 x 10(7) or 2 x 10(8) plaque forming units) vaccines were administered intramuscularly without adjuvants. Broadly reactive HIV-specific T cell immunity was stimulated by all doses of the vaccines administered, without significant differences between the high and low doses studied. The vaccines induced both CD4 and CD8 T cell responses to Gag, Pol, Env and Tat/Rev proteins, with CD4 T cell responses being greater in magnitude than CD8 T cell responses. The vaccine-induced T cell responses had significant cross-recognition of heterologous HIV-1 proteins from non-AE HIV-1 subtypes. In conclusion, these subtype AE HIV-1 DNA and rFPV vaccines were safe, induced broad T-cell immunity in macaques, and are suitable for progression into clinical trials.
Collapse
Affiliation(s)
- Robert De Rose
- Department of Microbiology and Immunology, University of Melbourne, Royal Parade, Vic. 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|