Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Virol ; 89(12): 6462-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25855741

ABSTRACT

UNLABELLED: An effective human immunodeficiency virus type 1 (HIV-1) vaccine must induce protective antibody responses, as well as CD4(+) and CD8(+) T cell responses, that can be effective despite extraordinary diversity of HIV-1. The consensus and mosaic immunogens are complete but artificial proteins, computationally designed to elicit immune responses with improved cross-reactive breadth, to attempt to overcome the challenge of global HIV diversity. In this study, we have compared the immunogenicity of a transmitted-founder (T/F) B clade Env (B.1059), a global group M consensus Env (Con-S), and a global trivalent mosaic Env protein in rhesus macaques. These antigens were delivered using a DNA prime-recombinant NYVAC (rNYVAC) vector and Env protein boost vaccination strategy. While Con-S Env was a single sequence, mosaic immunogens were a set of three Envs optimized to include the most common forms of potential T cell epitopes. Both Con-S and mosaic sequences retained common amino acids encompassed by both antibody and T cell epitopes and were central to globally circulating strains. Mosaics and Con-S Envs expressed as full-length proteins bound well to a number of neutralizing antibodies with discontinuous epitopes. Also, both consensus and mosaic immunogens induced significantly higher gamma interferon (IFN-γ) enzyme-linked immunosorbent spot assay (ELISpot) responses than B.1059 immunogen. Immunization with these proteins, particularly Con-S, also induced significantly higher neutralizing antibodies to viruses than B.1059 Env, primarily to tier 1 viruses. Both Con-S and mosaics stimulated more potent CD8-T cell responses against heterologous Envs than did B.1059. Both antibody and cellular data from this study strengthen the concept of using in silico-designed centralized immunogens for global HIV-1 vaccine development strategies. IMPORTANCE: There is an increasing appreciation for the importance of vaccine-induced anti-Env antibody responses for preventing HIV-1 acquisition. This nonhuman primate study demonstrates that in silico-designed global HIV-1 immunogens, designed for a human clinical trial, are capable of eliciting not only T lymphocyte responses but also potent anti-Env antibody responses.


Subject(s)
HIV-1/immunology , SAIDS Vaccines/immunology , Vaccination/methods , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Antigens, Viral/genetics , Antigens, Viral/immunology , Aspartate Aminotransferases , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Consensus Sequence , Enzyme-Linked Immunospot Assay , HIV Antibodies/blood , HIV-1/genetics , Humans , Interferon-gamma/metabolism , Macaca mulatta , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
2.
J Virol ; 87(24): 13589-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109227

ABSTRACT

Effective strategies are needed to block mucosal transmission of human immunodeficiency virus type 1 (HIV-1). Here, we address a crucial question in HIV-1 pathogenesis: whether infected donor mononuclear cells or cell-free virus plays the more important role in initiating mucosal infection by HIV-1. This distinction is critical, as effective strategies for blocking cell-free and cell-associated virus transmission may be different. We describe a novel ex vivo model system that utilizes sealed human colonic mucosa explants and demonstrate in both the ex vivo model and in vivo using the rectal challenge model in rhesus monkeys that HIV-1-infected lymphocytes can transmit infection across the mucosa more efficiently than cell-free virus. These findings may have significant implications for our understanding of the pathogenesis of mucosal transmission of HIV-1 and for the development of strategies to prevent HIV-1 transmission.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Intestinal Mucosa/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , Colon/virology , HIV-1/genetics , Humans , In Vitro Techniques , Macaca mulatta , Simian Immunodeficiency Virus/genetics
3.
Nature ; 441(7090): 239-43, 2006 May 11.
Article in English | MEDLINE | ID: mdl-16625206

ABSTRACT

A common viral immune evasion strategy involves mutating viral surface proteins in order to evade host neutralizing antibodies. Such immune evasion tactics have not previously been intentionally applied to the development of novel viral gene delivery vectors that overcome the critical problem of anti-vector immunity. Recombinant, replication-incompetent adenovirus serotype 5 (rAd5) vector-based vaccines for human immunodeficiency virus type 1 and other pathogens have proved highly immunogenic in preclinical studies but will probably be limited by the high prevalence of pre-existing anti-Ad5 immunity in human populations, particularly in the developing world. Here we show that rAd5 vectors can be engineered to circumvent anti-Ad5 immunity. We constructed novel chimaeric rAd5 vectors in which the seven short hypervariable regions (HVRs) on the surface of the Ad5 hexon protein were replaced with the corresponding HVRs from the rare adenovirus serotype Ad48. These HVR-chimaeric rAd5 vectors were produced at high titres and were stable through serial passages in vitro. HVR-chimaeric rAd5 vectors expressing simian immunodeficiency virus Gag proved comparably immunogenic to parental rAd5 vectors in naive mice and rhesus monkeys. In the presence of high levels of pre-existing anti-Ad5 immunity, the immunogenicity of HVR-chimaeric rAd5 vectors was not detectably suppressed, whereas the immunogenicity of parental rAd5 vectors was abrogated. These data demonstrate that functionally relevant Ad5-specific neutralizing antibodies are focused on epitopes located within the hexon HVRs. Moreover, these studies show that recombinant viral vectors can be engineered to circumvent pre-existing anti-vector immunity by removing key neutralizing epitopes on the surface of viral capsid proteins. Such chimaeric viral vectors may have important practical implications for vaccination and gene therapy.


Subject(s)
Adenoviridae/genetics , Adenoviridae/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Genetic Engineering , Genetic Vectors/genetics , Genetic Vectors/immunology , Adenoviridae/classification , Adenoviridae/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , DNA, Recombinant/genetics , Genetic Therapy , Macaca mulatta/immunology , Mice , Mice, Inbred C57BL , Neutralization Tests , Vaccines
4.
J Virol ; 84(24): 12782-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20881040

ABSTRACT

It has been suggested that vaccination prior to infection may direct the mutational evolution of human immunodeficiency virus type 1 (HIV-1) to a less fit virus, resulting in an attenuated course of disease. The present study was initiated to explore whether prior immunization might prevent the reversion of the virus to the wild-type form. Mamu-A*01 monkeys were vaccinated to generate a cytotoxic T-lymphocyte response to the immunodominant Gag p11C epitope and were then challenged with a cloned pathogenic CXCR4-tropic simian-human immunodeficiency virus (SHIV) expressing a mutant Gag p11C sequence (Δp11C SHIV). The epitopic and extraepitopic compensatory mutations introduced into gag of Δp11C SHIV resulted in attenuated replicative capacity and eventual reversions to the wild-type Gag p11C sequence in naïve rhesus monkeys. However, in vaccinated rhesus monkeys, no reversions of the challenge virus were observed, an effect that may have been a consequence of significantly decreased viral replication rather than a redirection of the mutational evolution of the virus. These findings highlight the multifactorial pressures that affect the evolution of primate immunodeficiency viruses.


Subject(s)
SAIDS Vaccines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/physiology , Virus Replication/genetics , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte/immunology , Flow Cytometry , Gene Products, gag/genetics , Histocompatibility Antigens Class I/immunology , Humans , Macaca mulatta , Molecular Sequence Data , Point Mutation , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Vaccination , Viral Load
5.
J Virol ; 83(19): 10028-35, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19641002

ABSTRACT

Emerging data suggest that a cytotoxic T-lymphocyte response against a diversity of epitopes confers greater protection against a human immunodeficiency virus/simian immunodeficiency virus infection than does a more focused response. To facilitate the creation of vaccine strategies that will generate cellular immune responses with the greatest breadth, it will be important to understand the mechanisms employed by the immune response to regulate the relative magnitudes of dominant and nondominant epitope-specific cellular immune responses. In this study, we generated dominant Gag p11C- and subdominant Env p41A-specific CD8(+) T-lymphocyte responses in Mamu-A*01(+) rhesus monkeys through vaccination with plasmid DNA and recombinant adenovirus encoding simian-human immunodeficiency virus (SHIV) proteins. Infection of vaccinated Mamu-A*01(+) rhesus monkeys with a SHIV Gag Deltap11C mutant virus generated a significantly increased expansion of the Env p41A-specific CD8(+) T-lymphocyte response in the absence of secondary Gag p11C-specific CD8(+) T-lymphocyte responses. These results indicate that the presence of the Gag p11C-specific CD8(+) T-lymphocyte response following virus challenge may exert suppressive effects on primed Env p41A-specific CD8(+) T-lymphocyte responses. These findings suggest that immunodomination exerted by dominant responses during SHIV infection may diminish the breadth of recall responses primed during vaccination.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Gene Products, gag/genetics , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Alleles , Animals , CD8-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Drug , Epitopes/chemistry , Flow Cytometry/methods , Gene Products, gag/chemistry , Macaca mulatta , Mutation , Peptides/chemistry , Point Mutation , Receptors, Antigen, T-Cell, alpha-beta/metabolism , SAIDS Vaccines/genetics , Simian Immunodeficiency Virus/genetics , Viral Load
6.
Science ; 369(6505): 812-817, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32434946

ABSTRACT

An understanding of protective immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical for vaccine and public health strategies aimed at ending the global coronavirus disease 2019 (COVID-19) pandemic. A key unanswered question is whether infection with SARS-CoV-2 results in protective immunity against reexposure. We developed a rhesus macaque model of SARS-CoV-2 infection and observed that macaques had high viral loads in the upper and lower respiratory tract, humoral and cellular immune responses, and pathologic evidence of viral pneumonia. After the initial viral clearance, animals were rechallenged with SARS-CoV-2 and showed 5 log10 reductions in median viral loads in bronchoalveolar lavage and nasal mucosa compared with after the primary infection. Anamnestic immune responses after rechallenge suggested that protection was mediated by immunologic control. These data show that SARS-CoV-2 infection induced protective immunity against reexposure in nonhuman primates.


Subject(s)
Betacoronavirus , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Betacoronavirus/immunology , Betacoronavirus/physiology , Bronchoalveolar Lavage Fluid/virology , COVID-19 , Coronavirus Infections/pathology , Coronavirus Infections/virology , Disease Models, Animal , Female , Immunity, Cellular , Immunity, Humoral , Immunologic Memory , Lung/immunology , Lung/pathology , Lung/virology , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/virology , Macaca mulatta , Male , Nasal Mucosa/virology , Pandemics , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Recurrence , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/immunology , Viral Load , Virus Replication
7.
J Clin Invest ; 114(9): 1334-42, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15520866

ABSTRACT

DCs are critical for priming adaptive immune responses to foreign antigens. However, the utility of harnessing these cells in vivo to optimize the immunogenicity of vaccines has not been fully explored. Here we investigate a novel vaccine approach that involves delivering synergistic signals that both recruit and expand DC populations at the site of antigen production. Intramuscular injection of an unadjuvanted HIV-1 envelope (env) DNA vaccine recruited few DCs to the injection site and elicited low-frequency, env-specific immune responses in mice. Coadministration of plasmids encoding the chemokine macrophage inflammatory protein-1alpha (MIP-1alpha) and the DC-specific growth factor fms-like tyrosine kinase 3 ligand with the DNA vaccine resulted in the recruitment, expansion, and activation of large numbers of DCs at the site of inoculation. Consistent with these findings, coadministration of these plasmid cytokines also markedly augmented DNA vaccine---elicited cellular and humoral immune responses and increased protective efficacy against challenge with recombinant vaccinia virus. These data suggest that the availability of mature DCs at the site of inoculation is a critical rate-limiting factor for DNA vaccine immunogenicity. Synergistic recruitment and expansion of DCs in vivo may prove a practical strategy for overcoming this limitation and potentiating immune responses to vaccines as well as other immunotherapeutic strategies.


Subject(s)
Dendritic Cells/cytology , Immunotherapy/methods , Vaccines, DNA , Animals , Cancer Vaccines , Chemokine CCL3 , Chemokine CCL4 , Chemotaxis , Cytokines/metabolism , DNA, Viral , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , HIV-1/genetics , Immunohistochemistry , Macrophage Inflammatory Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmids/metabolism , Time Factors , Vaccinia virus/genetics
8.
Nat Biotechnol ; 22(11): 1429-34, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15502816

ABSTRACT

In this study we extend tetramerization technology to T-cell receptors (TCRs). We identified TCR alpha beta pairs in the absence of accessory molecules, ensuring isolation of high-affinity TCRs that maintain stable binding characteristics after tetramerization. Subtle changes in cognate peptide levels bound to the class I molecule were accurately reflected by parallel changes in the mean fluorescence intensity of cells that bound TCR tetramers, allowing us to accurately assess the binding affinity of a panel of peptides to major histocompatibility complex (MHC) class I. Using a TCR tetramer specific for the Mamu-A(*)01 allele, we identified animals expressing this restricting class I allele from a large cohort of outbred rhesus macaques. TCR tetramers should facilitate analysis of the MHC-peptide interface and, more generally, the design of immunotherapeutics and vaccines.


Subject(s)
Fluorescence Polarization Immunoassay/methods , Genes, MHC Class I/immunology , Protein Engineering/methods , Protein Interaction Mapping/methods , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Antigen-Antibody Complex/analysis , Antigen-Antibody Reactions/immunology , Cells, Cultured , Dimerization , Genes, MHC Class I/genetics , Macaca mulatta , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics
9.
AIDS Res Hum Retroviruses ; 22(5): 445-52, 2006 May.
Article in English | MEDLINE | ID: mdl-16706622

ABSTRACT

Because of the importance of developing HIV vaccine strategies that generate cytotoxic T lymphocyte (CTL) responses with a maximal breadth of epitope recognition, we have explored a variety of novel strategies designed to overcome the usual propensity of CTLs to focus recognition on a limited number of dominant epitopes. In studies of rhesus monkeys expressing the Mamu-A*01 MHC class I allele, we show that variously configured multiepitope plasmid DNA vaccine constructs elicit CTL populations that do not evidence skewing of recognition to dominant epitopes. Nevertheless, repeated boosting of these vaccinated monkeys with different live recombinant vaccine vectors uncovers and amplifies the usual CTL epitope dominance hierarchy. Importantly, in vitro peptide stimulation of peripheral blood mononuclear cells from monkeys that have received only a multiepitope plasmid DNA priming immunization uncovers this dominance hierarchy. Therefore, the dominance hierarchy of the vaccine-elicited epitope-specific CTL populations is inherent in the T lymphocytes of the monkeys after initial exposure to epitope peptides, and the ultimate breadth of epitope recognition cannot be modified thereafter. This finding underscores the enormous challenge associated with increasing the breadth of CTL recognition through vaccination.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Immunodominant Epitopes/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/immunology , Alleles , Animals , In Vitro Techniques , Macaca mulatta , Peptides/immunology , Plasmids/genetics
10.
Clin Vaccine Immunol ; 21(10): 1385-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25080550

ABSTRACT

Live attenuated nonpathogenic Mycobacterium bovis bacillus Calmette-Guérin (BCG) mediates long-lasting immune responses, has been safely administered as a tuberculosis vaccine to billions of humans, and is affordable to produce as a vaccine vector. These characteristics make it very attractive as a human immunodeficiency virus (HIV) vaccine vector candidate. Here, we assessed the immunogenicity of recombinant BCG (rBCG) constructs with different simian immunodeficiency virus (SIV)gag expression cassettes as priming agents followed by a recombinant replication-incompetent New York vaccinia virus (NYVAC) boost in rhesus macaques. Unmutated rBCG constructs were used in comparison to mutants with gene deletions identified in an in vitro screen for augmented immunogenicity. We demonstrated that BCG-SIVgag is able to elicit robust transgene-specific priming responses, resulting in strong SIV epitope-specific cellular immune responses. While enhanced immunogenicity was sustained at moderate levels for >1 year following the heterologous boost vaccination, we were unable to demonstrate a protective effect after repeated rectal mucosal challenges with pathogenic SIVmac251. Our findings highlight the potential for rBCG vaccines to stimulate effective cross-priming and enhanced major histocompatibility complex class I presentation, suggesting that combining this approach with other immunogens may contribute to the development of effective vaccine regimens against HIV.


Subject(s)
Drug Carriers , Genetic Vectors , Mycobacterium bovis/genetics , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Animals , Gene Products, gag/genetics , Gene Products, gag/immunology , Immunity, Cellular , Macaca mulatta , Recombinant Proteins/genetics , Recombinant Proteins/immunology , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/genetics , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
11.
Nat Med ; 15(8): 873-5, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19620961

ABSTRACT

The immunologic basis for the potential enhanced HIV-1 acquisition in adenovirus serotype 5 (Ad5)-seropositive individuals who received the Merck recombinant Ad5 HIV-1 vaccine in the STEP study remains unclear. Here we show that baseline Ad5-specific neutralizing antibodies are not correlated with Ad5-specific T lymphocyte responses and that Ad5-seropositive subjects do not develop higher vector-specific cellular immune responses as compared with Ad5-seronegative subjects after vaccination. These findings challenge the hypothesis that activated Ad5-specific T lymphocytes were the cause of the potential enhanced HIV-1 susceptibility in the STEP study.


Subject(s)
AIDS Vaccines/immunology , AIDS Vaccines/therapeutic use , Acquired Immunodeficiency Syndrome/therapy , Adenoviridae/immunology , HIV-1/immunology , Acquired Immunodeficiency Syndrome/immunology , Antibodies, Viral/analysis , Antibodies, Viral/blood , Antibody Specificity/immunology , Humans , Immunization , Interferon-gamma/metabolism , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
12.
J Immunol ; 176(1): 319-28, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365424

ABSTRACT

Because the control of HIV-1 replication is largely dependent on CD8+ T lymphocyte responses specific for immunodominant viral epitopes, vaccine strategies that increase the breadth of dominant epitope-specific responses should contribute to containing HIV-1 spread. Developing strategies to elicit such broad immune responses will require an understanding of the mechanisms responsible for focusing CD8+ T lymphocyte recognition on a limited number of epitopes. To explore this biology, we identified cohorts of rhesus monkeys that expressed the MHC class I molecules Mamu-A*01, Mamu-A*02, or both, and assessed the evolution of their dominant epitope-specific CD8+ T lymphocyte responses (Gag p11C- and Tat TL8-specific in the Mamu-A*01+ and Nef p199RY-specific in the Mamu-A*02+ monkeys) following acute SIV infection. The Mamu-A*02+ monkeys that also expressed Mamu-A*01 exhibited a significant delay in the evolution of the CD8+ T lymphocyte responses specific for the dominant Mamu-A*02-restricted SIV epitope, Nef p199RY. This delay in kinetics was not due to differences in viral load kinetics or magnitude or in viral escape mutations, but was associated with the evolution of the Mamu-A*01-restricted CD8+ T lymphocyte responses to the highly dominant SIV epitopes Gag p11C and Tat TL8. Thus, the evolution of dominant epitope-specific CD8+ T lymphocyte responses can be suppressed by other dominant epitope-specific responses, and this immunodomination is important in determining the kinetics of dominant epitope-specific responses.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Immunodominant Epitopes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Amino Acid Sequence , Animals , Epitopes, T-Lymphocyte/genetics , Immunodominant Epitopes/genetics , Macaca mulatta , Molecular Sequence Data , Mutation , Polymerase Chain Reaction , Simian Immunodeficiency Virus/immunology
13.
J Virol ; 80(4): 1645-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16439521

ABSTRACT

Because the vaccine vectors currently being evaluated in human populations all have significant limitations in their immunogenicity, novel vaccine strategies are needed for the elicitation of cell-mediated immunity. The nonpathogenic, rapidly growing mycobacterium Mycobacterium smegmatis was engineered as a vector expressing full-length human immunodeficiency virus type 1 (HIV-1) HXBc2 envelope protein. Immunization of mice with recombinant M. smegmatis led to the expansion of major histocompatibility complex class I-restricted HIV-1 epitope-specific CD8(+) T cells that were cytolytic and secreted gamma interferon. Effector and memory T lymphocytes were elicited, and repeated immunization generated a stable central memory pool of virus-specific cells. Importantly, preexisting immunity to Mycobacterium bovis BCG had only a marginal effect on the immunogenicity of recombinant M. smegmatis. This mycobacterium may therefore be a useful vaccine vector.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Genes, env , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Mycobacterium smegmatis/genetics , Animals , Bacterial Vaccines/immunology , Cytotoxicity Tests, Immunologic , Female , Immunization , Immunologic Memory , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Mycobacterium bovis , Mycobacterium smegmatis/immunology , Vaccines, Synthetic/immunology
14.
Immunology ; 116(4): 443-53, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16313358

ABSTRACT

The development of successful vaccination strategies for eliciting cytotoxic T lymphocytes (CTLs) will be facilitated by the definition of strategies for subdividing CTLs into functionally distinct subpopulations. We assessed whether surface expression of a number of cell-surface proteins could be used to define functionally distinct subpopulations of memory CTLs in mice immunized with a recombinant vaccinia virus expressing human immunodeficiency virus (HIV)-1 envelope (Env). We found changes in cell-surface expression of CD11a, CD44, CD45RB, CD49d, CD54 and CD62L on Env-specific CD8(+) T cells that appeared to differentiate them from other CD8(+) T cells within 1 week to 1 month following immunization. Further, we saw an up-regulation of CD62L surface expression on Env-specific CD8(+) memory T cells several months after immunization. However, CD62L expression did not correlate with differences in the abilities of CTLs to proliferate or produce interferon gamma (IFN-gamma) and tumour necrosis factor alpha (TNF-alpha) in vitro in response to Env peptide stimulation. Moreover, the expression of CD62L did not allow differentiation of CTLs into subpopulations with distinct expansion kinetics in vivo after adoptive transfer into naïve mice and subsequent boosting of these mice with a recombinant adenovirus expressing HIV-1 Env. Therefore, the definition of memory CD8(+) T-cell subpopulations on the basis of CD62L expression in mice does not allow the delineation of functionally distinct CTL subpopulations.


Subject(s)
AIDS Vaccines/immunology , L-Selectin/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Biomarkers/metabolism , Cell Proliferation , Female , Immunologic Memory , Immunophenotyping , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Spleen/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Vaccination
15.
J Virol ; 79(15): 9694-701, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014931

ABSTRACT

The high prevalence of preexisting immunity to adenovirus serotype 5 (Ad5) in human populations will likely limit the immunogenicity and clinical utility of recombinant Ad5 (rAd5) vector-based vaccines for human immunodeficiency virus type 1 and other pathogens. A potential solution to this problem is to utilize rAd vaccine vectors derived from rare Ad serotypes such as Ad35 and Ad11. We have previously reported that rAd35 vectors were immunogenic in the presence of anti-Ad5 immunity, but the immunogenicity of heterologous rAd prime-boost regimens and the extent that cross-reactive anti-vector immunity may limit this approach have not been fully explored. Here we assess the immunogenicity of heterologous vaccine regimens involving rAd5, rAd35, and novel rAd11 vectors expressing simian immunodeficiency virus Gag in mice both with and without anti-Ad5 immunity. Heterologous rAd prime-boost regimens proved significantly more immunogenic than homologous regimens, as expected. Importantly, all regimens that included rAd5 were markedly suppressed by anti-Ad5 immunity. In contrast, rAd35-rAd11 and rAd11-rAd35 regimens elicited high-frequency immune responses both in the presence and in the absence of anti-Ad5 immunity, although we also detected clear cross-reactive Ad35/Ad11-specific humoral and cellular immune responses. Nevertheless, these data suggest the potential utility of heterologous rAd prime-boost vaccine regimens using vectors derived from rare human Ad serotypes.


Subject(s)
Adenoviruses, Human/immunology , Genetic Vectors/immunology , Reassortant Viruses/immunology , Viral Vaccines/immunology , Animals , Antibody Formation , Cross Reactions , Drug Evaluation, Preclinical , Gene Products, gag/genetics , Genetic Therapy , Immunity, Cellular , Immunization, Secondary , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Simian Immunodeficiency Virus/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage
16.
J Virol ; 79(10): 6554-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15858042

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.


Subject(s)
AIDS Vaccines/immunology , Genetic Vectors/genetics , HIV Infections/immunology , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Animals , Female , Gene Deletion , HIV Antibodies/blood , HIV Envelope Protein gp120/genetics , Immunization , Immunization, Secondary , Lethal Dose 50 , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/immunology , Virulence
17.
J Virol ; 79(22): 14161-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16254351

ABSTRACT

Preexisting immunity to adenovirus serotype 5 (Ad5) has been shown to suppress the immunogenicity of recombinant Ad5 (rAd5) vector-based vaccines for human immunodeficiency virus type 1 (HIV-1) in both preclinical studies and clinical trials. A potential solution to this problem is to utilize rAd vectors derived from rare Ad serotypes, such as Ad35. However, rAd35 vectors have appeared less immunogenic than rAd5 vectors in preclinical studies to date. In this study, we explore the hypothesis that the differences in immunogenicity between rAd5 and rAd35 vectors may be due in part to differences between the fiber proteins of these viruses. We constructed capsid chimeric rAd35 vectors containing the Ad5 fiber knob (rAd35k5) and compared the immunogenicities of rAd5, rAd35k5, and rAd35 vectors expressing simian immunodeficiency virus Gag and HIV-1 Env in mice and rhesus monkeys. In vitro studies demonstrated that rAd35k5 vectors utilized the Ad5 receptor CAR rather than the Ad35 receptor CD46. In vivo studies showed that rAd35k5 vectors were more immunogenic than rAd35 vectors in both mice and rhesus monkeys. These data suggest that the Ad5 fiber knob contributes substantially to the immunogenicity of rAd vectors. Moreover, these studies demonstrate that capsid chimeric rAd vectors can be constructed to combine beneficial immunologic and serologic properties of different Ad serotypes.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , Capsid Proteins/genetics , Viral Vaccines , Adenoviridae/classification , Adenoviridae/genetics , Animals , Epitopes/chemistry , Epitopes/immunology , Immunization , Macaca mulatta , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Serotyping , Virus Replication
18.
Nat Immunol ; 6(3): 247-52, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15685174

ABSTRACT

Viral escape from cytotoxic T lymphocytes (CTLs) can undermine immune control of human immunodeficiency virus 1. It is therefore important to assess the stability of viral mutations in CTL epitopes after transmission to naive hosts. Here we demonstrate the persistence of mutations in a dominant CTL epitope after transmission of simian immunodeficiency virus variants to major histocompatibility complex-matched rhesus monkeys. Transient reversions to wild-type sequences occurred and elicited CTLs specific for the wild-type epitope, resulting in immunological pressure that rapidly reselected the mutant viruses. These data suggest that mutations in dominant human immunodeficiency virus 1 CTL epitopes may accumulate in human populations with limited major histocompatibility complex heterogeneity by a mechanism involving dynamic CTL control of transiently reverted wild-type virus.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Mutation , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Epitopes, T-Lymphocyte/genetics , Humans , Immunodominant Epitopes/genetics , Macaca mulatta , Major Histocompatibility Complex/immunology , Simian Immunodeficiency Virus/pathogenicity
19.
J Immunol ; 174(8): 4753-60, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15814700

ABSTRACT

Production of IL-2 and IFN-gamma by CD4+ T lymphocytes is important for the maintenance of a functional immune system in infected individuals. In the present study, we assessed the cytokine production profiles of functionally distinct subsets of CD4+ T lymphocytes in rhesus monkeys infected with pathogenic or attenuated SIV/simian human immunodeficiency virus (SHIV) isolates, and these responses were compared with those in vaccinated monkeys that were protected from immunodeficiency following pathogenic SHIV challenge. We observed that preserved central memory CD4+ T lymphocyte production of SIV/SHIV-induced IL-2 was associated with disease protection following primate lentivirus infection. Persisting clinical protection in vaccinated and challenged monkeys is thus correlated with a preserved capacity of the peripheral blood central memory CD4+ T cells to express this important immunomodulatory cytokine.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV/immunology , Interleukin-2/biosynthesis , Simian Immunodeficiency Virus/immunology , AIDS Vaccines/pharmacology , Animals , CD28 Antigens/metabolism , Gene Products, gag , HIV/pathogenicity , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , Humans , Immunologic Memory , In Vitro Techniques , Interferon-gamma/biosynthesis , Macaca mulatta , RNA, Viral/blood , SAIDS Vaccines/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/biosynthesis , fas Receptor/metabolism
20.
J Immunol ; 174(11): 7179-85, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905562

ABSTRACT

The utility of recombinant adenovirus serotype 5 (rAd5) vector-based vaccines for HIV-1 and other pathogens will likely be limited by the high prevalence of pre-existing Ad5-specific neutralizing Abs (NAbs) in human populations. However, the immunodominant targets of Ad5-specific NAbs in humans remain poorly characterized. In this study, we assess the titers and primary determinants of Ad5-specific NAbs in individuals from both the United States and the developing world. Importantly, median Ad5-specific NAb titers were >10-fold higher in sub-Saharan Africa compared with the United States. Moreover, hexon-specific NAb titers were 4- to 10-fold higher than fiber-specific NAb titers in these cohorts by virus neutralization assays using capsid chimeric viruses. We next performed adoptive transfer studies in mice to evaluate the functional capacity of hexon- and fiber-specific NAbs to suppress the immunogenicity of a prototype rAd5-Env vaccine. Hexon-specific NAbs were remarkably efficient at suppressing Env-specific immune responses elicited by the rAd5 vaccine. In contrast, fiber-specific NAbs exerted only minimal suppressive effects on rAd5 vaccine immunogenicity. These data demonstrate that functionally significant Ad5-specific NAbs are directed primarily against the Ad5 hexon protein in both humans and mice. These studies suggest a potential strategy for engineering novel Ad5 vectors to evade dominant Ad5-specific NAbs.


Subject(s)
Adenoviruses, Human/immunology , Antibodies, Viral/physiology , Capsid Proteins/immunology , Genetic Vectors/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Adenoviruses, Human/genetics , Adult , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Dose-Response Relationship, Immunologic , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutralization Tests , Seroepidemiologic Studies , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL