Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 623996, 2021.
Article in English | MEDLINE | ID: mdl-33717130

ABSTRACT

The search for a preventive vaccine against HIV infection remains an ongoing challenge, indicating the need for novel approaches. Parainfluenza virus 5 (PIV5) is a paramyxovirus replicating in the upper airways that is not associated with any animal or human pathology. In animal models, PIV5-vectored vaccines have shown protection against influenza, RSV, and other human pathogens. Here, we generated PIV5 vaccines expressing HIV envelope (Env) and SIV Gag and administered them intranasally to macaques, followed by boosting with virus-like particles (VLPs) containing trimeric HIV Env. Moreover, we compared the immune responses generated by PIV5-SHIV prime/VLPs boost regimen in naïve vs a control group in which pre-existing immunity to the PIV5 vector was established. We demonstrate for the first time that intranasal administration of PIV5-based HIV vaccines is safe, well-tolerated and immunogenic, and that boosting with adjuvanted trimeric Env VLPs enhances humoral and cellular immune responses. The PIV5 prime/VLPs boost regimen induced robust and durable systemic and mucosal Env-specific antibody titers with functional activities including ADCC and neutralization. This regimen also induced highly polyfunctional antigen-specific T cell responses. Importantly, we show that diminished responses due to PIV5 pre-existing immunity can be overcome in part with VLP protein boosts. Overall, these results establish that PIV5-based HIV vaccine candidates are promising and warrant further investigation including moving on to primate challenge studies.


Subject(s)
AIDS Vaccines/administration & dosage , Gene Products, gag/administration & dosage , HIV-1/immunology , Immunogenicity, Vaccine , Parainfluenza Virus 5/immunology , Simian Immunodeficiency Virus/immunology , Virion/immunology , env Gene Products, Human Immunodeficiency Virus/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Cattle , Cell Line , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV-1/genetics , Host-Pathogen Interactions , Immunity, Cellular , Immunity, Humoral , Immunity, Mucosal , Macaca mulatta , Male , Nasal Mucosa/immunology , Nasal Mucosa/virology , Parainfluenza Virus 5/genetics , Simian Immunodeficiency Virus/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Virion/genetics , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
2.
Front Immunol ; 11: 1935, 2020.
Article in English | MEDLINE | ID: mdl-32983121

ABSTRACT

Studies have shown that vaccine vectors and route of immunization can differentially activate different arms of the immune system. However, the effects of different HIV vaccine immunogens on mucosal inflammation have not yet been studied. Because mucosal sites are the primary route of HIV infection, we evaluated the cervico-vaginal inflammatory cytokine and chemokine levels of Mauritian cynomolgus macaques following immunization and boost using two different SIV vaccine immunogens. The PCS vaccine delivers 12 20-amino acid peptides overlapping the 12 protease cleavage sites, and the Gag/Env vaccine delivers the full Gag and full Env proteins of simian immunodeficiency virus. We showed that the PCS vaccine prime and boosts induced short-lived, lower level increases of a few pro-inflammatory/chemotactic cytokines. In the PCS-vaccine group only the levels of MCP-1 were significantly increased above the baseline (P = 0.0078, Week 6; P = 0.0078, Week 17; P = 0.0234; Week 51) following multiple boosts. In contrast, immunizations with the Gag/Env vaccine persistently increased the levels of multiple cytokines/chemokines. In the Gag/Env group, higher than baseline levels were consistently observed for IL-8 (P = 0.0078, Week 16; P = 0.0078, Week 17; P = 0.0156, Week 52), IL-1ß (P = 0.0234, Week 16; P = 0.0156, Week 17; P = 0.0156, Week 52), and MIP-1α (P = 0.0313, Week 16; P = 0.0156, Week 17; P = 0.0313, Week 52). Over time, repeated boosts altered the relative levels of these cytokines between the Gag/Env and PCS vaccine group. 18 weeks after final boost with a higher dosage, IP-10 levels (P = 0.0313) in the Gag/Env group remained higher than baseline. Thus, the influence of vaccine immunogens on mucosal inflammation needs to be considered when developing and evaluating candidate HIV vaccines.


Subject(s)
Cervix Uteri/drug effects , Cytokines/metabolism , Gene Products, env/administration & dosage , Gene Products, gag/administration & dosage , Inflammation Mediators/metabolism , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vagina/drug effects , Animals , Cervix Uteri/immunology , Cervix Uteri/metabolism , Female , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, env/toxicity , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, gag/toxicity , Macaca fascicularis , Mucous Membrane/drug effects , Mucous Membrane/immunology , Mucous Membrane/metabolism , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , SAIDS Vaccines/toxicity , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Time Factors , Vaccination , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/toxicity , Vagina/immunology , Vagina/metabolism
3.
Bing Du Xue Bao ; 32(2): 170-8, 2016 Mar.
Article in Chinese | MEDLINE | ID: mdl-27396160

ABSTRACT

Therapeutic HIV vaccine was considered as a hopeful curative method for AIDS patients. However, there is still no suitable HIV animal model for vaccine study since the difference in the immune system between human and animals. To evaluate the therapeutic effect of combined immunization strategy with multiple vector vaccines in macaque models. Plasmid DNA, recombinant Ad5 and MVA vaccines which expressing SIV gag and env genes were constructed. Sequential and repeated immune strategy were applied to immunize mice with these three vaccines. Cellular immune responses in mice immunized with these three vaccines were measured by ELISPOT test in vitro and CTL assay in vivo. The results were analyzed and compared with different antigen combination, order of vaccines and intervals to choose a suitable immunization strategy for macaque immunization in future. It indicated that strong SIV-Gag/Env-specific cellular immune responses were induced by these three vector vaccines. It laid a foundation for evaluating the therapeutic effect of combined immunization strategy with multiple vector vaccines in SIV infected macaque models.


Subject(s)
AIDS Vaccines/immunology , Gene Products, env/immunology , Gene Products, gag/immunology , HIV Infections/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antibodies, Viral/immunology , Female , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , HIV Infections/prevention & control , HIV Infections/virology , Humans , Immunization , Mice , Mice, Inbred BALB C , Simian Immunodeficiency Virus/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
4.
Virology ; 476: 405-412, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25591175

ABSTRACT

We reported previously on a vaccine approach that conferred apparent sterilizing immunity to SIVsmE660. The vaccine regimen employed a prime-boost using vectors based on recombinant vesicular stomatitis virus (VSV) and an alphavirus replicon expressing either SIV Gag or SIV Env. In the current study, we tested the ability of vectors expressing only the SIVsmE660 Env protein to protect macaques against the same high-dose mucosal challenge. Animals developed neutralizing antibody levels comparable to or greater than seen in the previous vaccine study. When the vaccinated animals were challenged with the same high-dose of SIVsmE660, all became infected. While average peak viral loads in animals were slightly lower than those of previous controls, the viral set points were not significantly different. These data indicate that Gag, or the combination of Gag and Env are required for the generation of apparent sterilizing immunity to the SIVsmE660 challenge.


Subject(s)
AIDS Vaccines/immunology , Gene Products, env/immunology , Gene Products, gag/immunology , Mucous Membrane/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , HIV/genetics , HIV/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , Humans , Macaca mulatta , Mucous Membrane/virology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics
5.
Wei Sheng Wu Xue Bao ; 54(3): 352-8, 2014 Mar 04.
Article in Chinese | MEDLINE | ID: mdl-24984528

ABSTRACT

OBJECTIVE: To study the immunogenicity of co-expression of p30, one of a group specific antigens, and glycoprotein gp90 genes of Reticuloendotheliosis virus (REV). METHODS: p30 and gp90 genes were amplified with the template of plasmid pPB101 containing the whole sequence of spleen necrosis virus (SNV), and cloned into pET-28a(+) vector. The positive clone pET-p30-gp90 was identified by enzyme analysis and sequencing. The co-expressed protein p30-gp90 was confirmed by SDS-PAGE and Western blot analysis. After quantitation, the purified protein p30-gp90 was immunized to Balb/c mice three times to prepare the p30-gp90 specific anti-serum. Afterwards, we detected the REV infected Chick Embryo Fibroblasts (CEF) by Immunofluorescence assay (IFA) with the prepared anti-serum. RESULTS: The p30-gp90 was co-expressed efficiently by SDS-PAGE and Western blot analysis. The anti-serum of p30-gp90 reacts with REV infected CEF in IFA. CONCLUSION: The expressed protein of p30-gp90 keeps good immunogenicity.


Subject(s)
Gene Products, env/immunology , Gene Products, gag/immunology , Poultry Diseases/prevention & control , Recombinant Fusion Proteins/immunology , Reticuloendotheliosis virus/immunology , Retroviridae Infections/veterinary , Animals , Chick Embryo , Chickens , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , Humans , Mice , Mice, Inbred BALB C , Poultry Diseases/immunology , Poultry Diseases/virology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Reticuloendotheliosis virus/genetics , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Retroviridae Infections/virology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
6.
J Immunol ; 190(8): 4103-15, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23509365

ABSTRACT

Prime-boost immunization with heterologous vaccines elicits potent cellular immunity. In this study, we assessed the influence of various TLR ligands on SIV Gag-specific T cell immunity and protection following prime-boost immunization. Rhesus macaques (RMs) were primed with SIV Gag protein emulsified in Montanide ISA51 with or without TLR3 (polyinosinic-polycytidylic acid [poly-IC]), TLR4 (monophosphoryl lipid A), TLR7/8 (3M-012), TLR9 (CpG), or TLR3 (poly-IC) combined with TLR7/8 ligands, then boosted with replication defective adenovirus 5 expressing SIV Gag (rAd5-Gag). After priming, RMs that received SIV Gag protein plus poly-IC developed significantly higher frequencies of SIV Gag-specific CD4(+) Th1 responses in blood and bronchoalveolar lavage (BAL) fluid lymphocytes compared with all other adjuvants, and low-level SIV Gag-specific CD8(+) T cell responses. After the rAd5-Gag boost, the magnitude and breadth of SIV Gag-specific CD8(+) T cell responses were significantly increased in RM primed with SIV Gag protein plus poly-IC, with or without the TLR7/8 ligand, or CpG. However, the anamnestic, SIV Gag-specific CD8(+) T cell response to SIVmac251 challenge was not significantly enhanced by SIV Gag protein priming with any of the adjuvants. In contrast, the anamnestic SIV Gag-specific CD4(+) T cell response in BAL was enhanced by SIV Gag protein priming with poly-IC or CpG, which correlated with partial control of early viral replication after SIVmac251 challenge. These results demonstrate that prime-boost vaccination with SIV Gag protein/poly-IC improves magnitude, breadth, and durability of CD4(+) T cell immune responses, which could have a role in the control of SIV viral replication.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Gene Products, gag/administration & dosage , Poly I-C/administration & dosage , Receptors, Antigen, T-Cell/physiology , T-Lymphocyte Subsets/immunology , Adjuvants, Immunologic/therapeutic use , Animals , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Gene Products, gag/immunology , Macaca mulatta , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Mannitol/therapeutic use , Oleic Acids/administration & dosage , Oleic Acids/therapeutic use , Poly I-C/therapeutic use , Simian Immunodeficiency Virus/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology
7.
J Immunol ; 190(6): 2720-35, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23390298

ABSTRACT

Recombinant adenoviral vectors (rAds) are the most potent recombinant vaccines for eliciting CD8(+) T cell-mediated immunity in humans; however, prior exposure from natural adenoviral infection can decrease such responses. In this study we show low seroreactivity in humans against simian- (sAd11, sAd16) or chimpanzee-derived (chAd3, chAd63) compared with human-derived (rAd5, rAd28, rAd35) vectors across multiple geographic regions. We then compared the magnitude, quality, phenotype, and protective capacity of CD8(+) T cell responses in mice vaccinated with rAds encoding SIV Gag. Using a dose range (1 × 10(7)-10(9) particle units), we defined a hierarchy among rAd vectors based on the magnitude and protective capacity of CD8(+) T cell responses, from most to least, as: rAd5 and chAd3, rAd28 and sAd11, chAd63, sAd16, and rAd35. Selection of rAd vector or dose could modulate the proportion and/or frequency of IFN-γ(+)TNF-α(+)IL-2(+) and KLRG1(+)CD127(-)CD8(+) T cells, but strikingly ∼30-80% of memory CD8(+) T cells coexpressed CD127 and KLRG1. To further optimize CD8(+) T cell responses, we assessed rAds as part of prime-boost regimens. Mice primed with rAds and boosted with NYVAC generated Gag-specific responses that approached ∼60% of total CD8(+) T cells at peak. Alternatively, priming with DNA or rAd28 and boosting with rAd5 or chAd3 induced robust and equivalent CD8(+) T cell responses compared with prime or boost alone. Collectively, these data provide the immunologic basis for using specific rAd vectors alone or as part of prime-boost regimens to induce CD8(+) T cells for rapid effector function or robust long-term memory, respectively.


Subject(s)
Adenoviridae/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Gene Products, gag/immunology , Genetic Vectors/administration & dosage , HIV-1/immunology , Quality Assurance, Health Care , Simian Immunodeficiency Virus/immunology , Adenoviridae/genetics , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Disease Models, Animal , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/therapeutic use , Gene Products, gag/administration & dosage , Gene Products, gag/therapeutic use , Genetic Vectors/immunology , Genetic Vectors/therapeutic use , HEK293 Cells , HIV-1/genetics , Humans , Immunophenotyping/methods , Immunophenotyping/standards , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pan troglodytes , Quality Assurance, Health Care/standards , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Simian Immunodeficiency Virus/genetics
8.
J Immunol ; 188(2): 714-23, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22174446

ABSTRACT

The majority of HIV infections occur via mucosal transmission. Vaccines that induce memory T and B cells in the female genital tract may prevent the establishment and systemic dissemination of HIV. We tested the immunogenicity of a vaccine that uses human papillomavirus (HPV)-based gene transfer vectors, also called pseudovirions (PsVs), to deliver SIV genes to the vaginal epithelium. Our findings demonstrate that this vaccine platform induces gene expression in the genital tract in both cynomolgus and rhesus macaques. Intravaginal vaccination with HPV16, HPV45, and HPV58 PsVs delivering SIV Gag DNA induced Gag-specific Abs in serum and the vaginal tract, and T cell responses in blood, vaginal mucosa, and draining lymph nodes that rapidly expanded following intravaginal exposure to SIV(mac251.) HPV PsV-based vehicles are immunogenic, which warrant further testing as vaccine candidates for HIV and may provide a useful model to evaluate the benefits and risks of inducing high levels of SIV-specific immune responses at mucosal sites prior to SIV infection.


Subject(s)
DNA, Viral/administration & dosage , Gene Products, gag/genetics , Gene Transfer Techniques , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/genetics , Simian Immunodeficiency Virus/genetics , Vagina/immunology , Virion/genetics , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Animals , DNA, Viral/immunology , Female , Gene Products, gag/administration & dosage , Gene Products, gag/immunology , HEK293 Cells , Humans , Immunity, Mucosal/genetics , Luminescent Proteins/administration & dosage , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Macaca fascicularis , Macaca mulatta , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Vagina/metabolism , Vagina/virology , Virion/immunology , Red Fluorescent Protein
9.
J Virol ; 86(4): 2239-50, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22156519

ABSTRACT

The Step Trial showed that the MRKAd5 HIV-1 subtype B Gag/Pol/Nef vaccine did not protect men from HIV infection or reduce setpoint plasma viral RNA (vRNA) levels but, unexpectedly, it did modestly enhance susceptibility to HIV infection in adenovirus type 5 (Ad5)-seropositive, uncircumcised men. As part of the process to understand the results of the Step Trial, we designed a study to determine whether rhesus macaques chronically infected with a host-range mutant Ad5 (Ad5hr) and then immunized with a replication defective Ad5 SIVmac239 Gag/Pol/Nef vaccine were more resistant or susceptible to SIV infection than unimmunized rhesus macaques challenged with a series of escalating dose penile exposures to SIVmac 251. The Ad5 SIV vaccine induced CD8(+) T cell responses in 70% of the monkeys, which is similar to the proportion of humans that responded to the vaccine in the Step Trial. However, the vaccine did not protect vaccinated animals from penile SIV challenge. At the lowest SIV exposure dose (10(3) 50% tissue culture infective doses), 2 of 9 Ad5-seropositive animals immunized with the Ad5 SIV vaccine became infected compared to 0 of 34 animals infected in the other animal groups (naive animals, Ad5-seropositive animals immunized with the empty Ad5 vector, Ad5-seronegative animals immunized with the Ad5 SIV vaccine, and Ad5-seronegative animals immunized with the empty Ad5 vector). Penile exposure to more concentrated virus inocula produced similar rates of infection in all animal groups. Although setpoint viral loads were unaffected in Step vaccinees, the Ad5 SIV-immunized animals had significantly lower acute-phase plasma vRNA levels compared to unimmunized animals. Thus, the results of the nonhuman primate (NHP) study described here recapitulate the lack of protection against HIV acquisition seen in the Step Trial and suggest a greater risk of infection in the Ad5-seropositive animals immunized with the Ad5 SIV vaccine. Further studies are necessary to confirm the enhancement of virus acquisition and to discern associated mechanisms.


Subject(s)
AIDS Vaccines/immunology , Gene Products, env/immunology , Gene Products, gag/immunology , Gene Products, nef/immunology , HIV Infections/prevention & control , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Animals , Antibodies, Viral/immunology , Defective Viruses/genetics , Defective Viruses/physiology , Disease Models, Animal , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , Gene Products, nef/administration & dosage , Gene Products, nef/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , HIV/genetics , HIV/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Humans , Immunization , Macaca mulatta , Male , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics
10.
J Virol ; 85(23): 12708-20, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21900170

ABSTRACT

Recombinant strains of replication-competent rhesus monkey rhadinovirus (RRV) were constructed in which strong promoter/enhancer elements were used to drive expression of simian immunodeficiency virus (SIV) Env or Gag or a Rev-Tat-Nef fusion protein. Cultured rhesus monkey fibroblasts infected with each recombinant strain were shown to express the expected protein. Three RRV-negative and two RRV-positive rhesus monkeys were inoculated intravenously with a mixture of these three recombinant RRVs. Expression of SIV Gag was readily detected in lymph node biopsy specimens taken at 3 weeks postimmunization. Impressive anti-SIV cellular immune responses were elicited on the basis of major histocompatibility complex (MHC) tetramer staining and gamma interferon enzyme-linked immunospot (ELISPOT) assays. Responses were much greater in magnitude in the monkeys that were initially RRV negative but were still readily detected in the two monkeys that were naturally infected with RRV at the time of immunization. By 3 weeks postimmunization, responses measured by MHC tetramer staining in the two Mamu-A*01(+) RRV-negative monkeys reached 9.3% and 13.1% of all CD8(+) T cells in peripheral blood to the Gag CM9 epitope and 2.3% and 7.3% of all CD8(+) T cells in peripheral blood to the Tat SL8 epitope. Virus-specific CD8(+) T cell responses persisted at high levels up to the time of challenge at 18 weeks postimmunization, and responding cells maintained an effector memory phenotype. Despite the ability of the RRVenv recombinant to express high levels of Env in cultured cells, and despite the appearance of strong anti-RRV antibody responses in immunized monkeys, anti-Env antibody responses were below our ability to detect them. Immunized monkeys, together with three unimmunized controls, were challenged intravenously with 10 monkey infectious doses of SIVmac239. All five immunized monkeys and all three controls became infected with SIV, but peak viral loads were 1.2 to 3.0 log(10) units lower and chronic-phase viral loads were 1.0 to 3.0 log(10) units lower in immunized animals than the geometric mean of unimmunized controls. These differences were statistically significant. Anti-Env antibody responses following challenge indicated an anamnestic response in the vaccinated monkeys. These findings further demonstrate the potential of recombinant herpesviruses as preventive vaccines for AIDS. We hypothesize that this live, replication-competent, persistent herpesvirus vector could match, or come close to matching, live attenuated strains of SIV in the degree of protection if the difficulty with elicitation of anti-Env antibody responses can be overcome.


Subject(s)
Gammaherpesvirinae/immunology , Herpesviridae Infections/metabolism , Macaca mulatta/immunology , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Viral/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gammaherpesvirinae/genetics , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, nef/genetics , Gene Products, nef/immunology , Genetic Vectors , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Humans , Immunity, Cellular , Immunoenzyme Techniques , Kidney/cytology , Kidney/metabolism , Kidney/virology , Macaca mulatta/genetics , Macaca mulatta/virology , Neutralization Tests , Plasmids , Recombination, Genetic , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Vaccination , Viral Load , Virus Replication
11.
Retrovirology ; 8: 57, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21752277

ABSTRACT

BACKGROUND: HIV infection causes a qualitative and quantitative loss of CD4+ T cell immunity. The institution of anti-retroviral therapy (ART) restores CD4+ T cell responses to many pathogens, but HIV-specific responses remain deficient. Similarly, therapeutic immunization with HIV antigens of chronically infected, ART treated subjects results in poor induction of HIV-specific CD4 responses. In this study, we used a macaque model of ART treatment during chronic infection to study the virologic consequences of SIV antigen stimulation in lymph nodes early after immunization. Rhesus CMV (RhCMV) seropositive, Mamu A*01 positive rhesus macaques were chronically infected with SIVmac251 and treated with ART. The immune and viral responses to SIV gag and RhCMV pp65 antigen immunization in draining lymph nodes and peripheral blood were analyzed. Animals were immunized on contralateral sides with SIV gag and RhCMV pp65 encoding plasmids, which allowed lymph nodes draining each antigen to be obtained at the same time from the same animal for direct comparison. RESULTS: We observed that both SIV and RhCMV immunizations stimulated transient antigen-specific T cell responses in draining lymph nodes. The RhCMV-specific responses were potent and sustained (50 days post-immunization) in the periphery, while the SIV-specific responses were transient and extinguished quickly. The SIV antigen stimulation selectively induced transient SIV replication in draining lymph nodes. CONCLUSIONS: The data are consistent with a model whereby viral replication in response to SIV antigen stimulation limits the generation of SIV antigen-specific responses and suggests a potential mechanism for the early loss and poor HIV-specific CD4+ T cell response observed in HIV-infected individuals.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , HIV Infections/immunology , Lymph Nodes/virology , Macaca mulatta/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Virus Replication , Animals , Antibodies, Viral , CD4-Positive T-Lymphocytes/virology , Gene Products, gag/administration & dosage , Gene Products, gag/immunology , HIV Infections/drug therapy , HIV Infections/virology , Humans , Immunization , Immunotherapy, Active , Lymph Nodes/immunology , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology
12.
Vaccine ; 29(34): 5611-22, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21693155

ABSTRACT

We sought to induce primate immunodeficiency virus-specific cellular and neutralizing antibody (nAb) responses in rhesus macaques (RM) through a bimodal vaccine approach. RM were immunized intragastrically (i.g.) with the live-attenuated Listeria monocytogenes (Lm) vector Lmdd-BdopSIVgag encoding SIVmac239 gag. SIV Gag-specific cellular responses were boosted by intranasal and intratracheal administration of replication-competent adenovirus (Ad5hr-SIVgag) encoding the same gag. To broaden antiviral immunity, the RM were immunized with multimeric HIV clade C (HIV-C) gp160 and HIV Tat. SIV Gag-specific cellular immune responses and HIV-1 nAb developed in some RM. The animals were challenged intrarectally with five low doses of R5 SHIV-1157ipEL-p, encoding a heterologous HIV-C Env (22.1% divergent to the Env immunogen). All five controls became viremic. One out of ten vaccinees was completely protected and another had low peak viremia. Sera from the completely and partially protected RM neutralized the challenge virus > 90%; these RM also had strong SIV Gag-specific proliferation of CD8⁺ T cells. Peak and area under the curve of plasma viremia (during acute phase) among vaccinees was lower than for controls, but did not attain significance. The completely protected RM showed persistently low numbers of the α4ß7-expressing CD4⁺ T cells; the latter have been implicated as preferential virus targets in vivo. Thus, vaccine-induced immune responses and relatively lower numbers of potential target cells were associated with protection.


Subject(s)
Adenoviridae/immunology , Gene Products, gag/immunology , HIV Envelope Protein gp160/immunology , Listeria monocytogenes/immunology , tat Gene Products, Human Immunodeficiency Virus/immunology , Adenoviridae/genetics , Animals , Antibodies, Neutralizing/blood , Antigens, Viral/genetics , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Enzyme-Linked Immunospot Assay , Gene Products, gag/administration & dosage , HIV Envelope Protein gp160/administration & dosage , HIV Infections/prevention & control , HIV-1/immunology , Immunity, Mucosal , Immunization, Secondary , Interferon-gamma/analysis , Listeria monocytogenes/genetics , Macaca mulatta/immunology , Macaca mulatta/virology , Simian Immunodeficiency Virus/immunology , Vaccination , Viral Load , Viremia/immunology , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
13.
J Immunol ; 186(12): 6914-24, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21576505

ABSTRACT

Polyvalent mosaic HIV immunogens offer a potential solution for generating vaccines that can elicit immune responses against genetically diverse viruses. However, it is unclear whether key T cell epitopes can be processed and presented from these synthetic Ags and recognized by epitope-specific human T cells. In this study, we tested the ability of mosaic HIV immunogens expressed by recombinant, replication-incompetent adenovirus serotype 26 vectors to process and present major HIV clade B and clade C CD8 T cell epitopes in human cells. A bivalent mosaic vaccine expressing HIV Gag sequences was used to transduce PBMCs from 12 HIV-1-infected individuals from the United States and 10 HIV-1-infected individuals from South Africa; intracellular cytokine staining, together with tetramer staining, was used to assess the ability of mosaic Gag Ags to stimulate pre-existing memory responses compared with natural clade B and C vectors. Mosaic Gag Ags expressed all eight clade B epitopes tested in 12 United States subjects and all 5 clade C epitopes tested in 10 South African subjects. Overall, the magnitude of cytokine production induced by stimulation with mosaic Ags was comparable to clade B and clade C Ags tested, but the mosaic Ags elicited greater cross-clade recognition. Additionally, mosaic Ags induced HIV-specific CD4 T cell responses. Our studies demonstrate that mosaic Ags express major clade B and clade C viral T cell epitopes in human cells, as well as support the evaluation of mosaic HIV-1 vaccines in humans.


Subject(s)
AIDS Vaccines/immunology , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Products, gag/immunology , AIDS Vaccines/genetics , Adenoviridae/genetics , Cross Reactions/immunology , Gene Products, gag/administration & dosage , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HIV-1/immunology , Humans , South Africa , Species Specificity , United States
14.
J Virol ; 85(13): 6442-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490092

ABSTRACT

Major histocompatibility complex (MHC) molecules expressed on the surface of human immunodeficiency virus (HIV) are potential targets for neutralizing antibodies. Since MHC molecules are polymorphic, nonself MHC can also be immunogenic. We have used combinations of novel recombinant HLA class I and II and HIV/simian immunodeficiency virus (SIV) antigens, all linked to dextran, to investigate whether they can elicit protective immunity against heterologous simian/human immunodeficiency virus (SHIV) challenge in rhesus macaques. Three groups of animals were immunized with HLA (group 1, n = 8), trimeric YU2 HIV type 1 (HIV-1) gp140 and SIV p27 (HIV/SIV antigens; group 2, n = 8), or HLA plus HIV/SIV antigens (group 3, n = 8), all with Hsp70 and TiterMax Gold adjuvant. Another group (group 4, n = 6) received the same vaccine as group 3 without TiterMax Gold. Two of eight macaques in group 3 were completely protected against intravenous challenge with 18 50% animal infective doses (AID(50)) of SHIV-SF162P4/C grown in human cells expressing HLA class I and II lineages represented in the vaccine, while the remaining six macaques showed decreased viral loads compared to those in unimmunized animals. Complement-dependent neutralizing activity in serum and high levels of anti-HLA antibodies were elicited in groups 1 and 3, and both were inversely correlated with the plasma viral load at 2 weeks postchallenge. Antibody-mediated protection was strongly supported by the fact that transfer of pooled serum from the two challenged but uninfected animals protected two naïve animals against repeated low-dose challenge with the same SHIV stock. This study demonstrates that immunization with recombinant HLA in combination with HIV-1 antigens might be developed into an alternative strategy for a future AIDS vaccine.


Subject(s)
AIDS Vaccines/administration & dosage , Gene Products, gag/administration & dosage , HIV Infections/prevention & control , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class I/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , env Gene Products, Human Immunodeficiency Virus/administration & dosage , Animals , Female , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV Infections/immunology , HIV-1/immunology , HIV-1/pathogenicity , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunization , Macaca mulatta/immunology , Molecular Sequence Data , Recombination, Genetic , SAIDS Vaccines/administration & dosage , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Treatment Outcome , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
15.
J Immunol ; 185(11): 7097-106, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21041730

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.


Subject(s)
DNA, Viral/immunology , Genetic Vectors/immunology , Immunity, Cellular , Immunization, Secondary/methods , Lactation/immunology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/immunology , SAIDS Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , DNA, Recombinant/administration & dosage , DNA, Recombinant/immunology , DNA, Viral/administration & dosage , DNA, Viral/genetics , Female , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, pol/administration & dosage , Gene Products, pol/genetics , Gene Products, pol/immunology , Genetic Vectors/administration & dosage , Lactation/genetics , Macaca mulatta , Mammary Glands, Animal/metabolism , Poxviridae/genetics , Poxviridae/immunology , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/transmission , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
16.
J Virol ; 83(21): 10963-74, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19706700

ABSTRACT

AIDS vaccination has a pressing need for more potent vaccination vectors capable of eliciting strong, diversified, and long-lasting cellular immune responses against human immunodeficiency virus (HIV). Lentiviral vectors have demonstrated efficiency not only as gene delivery vehicles for gene therapy applications but also as vaccination tools. This is likely due to their ability to transduce nondividing cells, including dendritic cells, enabling sustained endogenous antigen presentation and thus the induction of high proportions of specific cytotoxic T cells and long-lasting memory T cells. We show in a first proof-of-concept pilot study that a prime/boost vaccination strategy using lentiviral vectors pseudotyped with a glycoprotein G from two non-cross-reactive vesicular stomatitis virus serotypes elicited robust and broad cellular immune responses against the vector-encoded antigen, simian immunodeficiency virus (SIV) GAG, in cynomolgus macaques. Vaccination conferred strong protection against a massive intrarectal challenge with SIVmac251, as evidenced both by the reduction of viremia at the peak of acute infection (a mean of over 2 log(10) fold reduction) and by the full preservation of the CD28(+) CD95(+) memory CD4(+) T cells during the acute phase, a strong correlate of protection against pathogenesis. Although vaccinees continued to display lower viremia than control macaques during the early chronic phase, these differences were not statistically significant by day 50 postchallenge. A not-optimized SIV GAG antigen was chosen to show the strong potential of the lentiviral vector system for vaccination. Given that a stronger protection can be anticipated from a modern HIV-1 antigen design, gene transfer vectors derived from HIV-1 appear as promising candidates for vaccination against HIV-1 infection.


Subject(s)
AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome , Aspartic Acid Endopeptidases , Genetic Vectors , Lentivirus , Simian Immunodeficiency Virus/immunology , Vaccination , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/prevention & control , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/immunology , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , Gene Products, gag/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Lentivirus/genetics , Lentivirus/immunology , Macaca/immunology , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Male , Pilot Projects , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Viremia
17.
J Virol ; 83(20): 10840-3, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19640980

ABSTRACT

Recent findings suggest that most sexual transmission of human immunodeficiency virus type 1 (HIV-1) occurs during the acute phase of infection when viral replication is most intense. Here, we show that vaccine-elicited cellular immune responses can significantly reduce simian immunodeficiency virus levels in the semen during the period of primary infection in monkeys. A vaccine that decreases the quantity of HIV-1 in the semen of males during primary infection might decrease HIV-1 transmission in human populations and therefore affect the spread of AIDS.


Subject(s)
SAIDS Vaccines/immunology , Semen/virology , Simian Immunodeficiency Virus/isolation & purification , T-Lymphocytes/immunology , Animals , Gene Products, gag/administration & dosage , Gene Products, gag/immunology , Gene Products, pol/administration & dosage , Gene Products, pol/immunology , Humans , Lymphocyte Activation , Macaca mulatta , Male , RNA, Viral/analysis , RNA, Viral/blood , Reverse Transcriptase Polymerase Chain Reaction , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Vaccination , Viral Load
18.
Vaccine ; 27(34): 4591-600, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19538997

ABSTRACT

RNA transcripts of the B7 family molecule (CD80) are diminished in blood leukocytes from animals clinically affected with Visna/Maedi virus (VMV) infection. This work investigates whether the use of B7 genes enhances immune responses and protection in immunization-challenge approaches. Sheep were primed by particle-mediated epidermal bombardment with VMV gag and env gene recombinant plasmids together with plasmids encoding both CD80 and CD86 or CD80 alone, boosted with gag and env gene recombinant modified vaccinia Ankara virus and challenged intratracheally with VMV. Immunization in the presence of one or both of the B7 genes resulted in CD4+ T cell activation and antibody production (before and after challenge, respectively), but only immunization with CD80 and CD86 genes together, and not CD80 alone, resulted in a reduced number of infected animals and increased early transient cytotoxic T lymphocytes (CTL) responses. Post-mortem analysis showed an immune activation of lymphoid tissue in challenge-target organs in those animals that had received B7 genes compared to unvaccinated animals. Thus, the inclusion of B7 genes helped to enhance early cellular responses and protection (diminished proportion of infected animals) against VMV infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , B7-1 Antigen/administration & dosage , Pneumonia, Progressive Interstitial, of Sheep/prevention & control , Vaccines, DNA/immunology , Viral Vaccines/immunology , Visna-maedi virus/immunology , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Viral/blood , B7-1 Antigen/genetics , B7-1 Antigen/pharmacology , B7-2 Antigen/administration & dosage , B7-2 Antigen/genetics , B7-2 Antigen/pharmacology , CD4-Positive T-Lymphocytes/immunology , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , Genetic Vectors , Immunization, Secondary/methods , Male , Sheep , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/genetics , Visna-maedi virus/genetics
19.
Vaccine ; 27(4): 530-41, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19026705

ABSTRACT

Influenza-pseudotyped Gag virus-like particles (VLPs) were produced via the expression of influenza hemagglutinin (HA), neuraminidase (NA) and the murine leukemia virus Gag product in the baculovirus-insect cell expression system. Hemagglutination specific activities of sucrose gradient-purified VLPs were similar to those of egg-grown influenza viruses but particle morphologies were gamma retrovirus-like in the form of consistent 100nm spheres. Immunization of mice and ferrets demonstrated robust immunogenicity and protection from challenge with no measurable morbidity. Ferret data were striking in that immunization with H5N1 VLPs representing either A/Vietnam/1203/04 or A/Indonesia/5/05 resulted in solid protection against highly pathogenic A/Vietnam/1203/04 challenge with no detectable virus in the upper respiratory tract post-challenge in either group. H1N1 VLP immunization of ferrets resulted in partial protection against H5N1 challenge with markedly accelerated virus clearance from the upper respiratory tract relative to controls. The immunogenicity of influenza-pseudotyped VLPs was not dependent on the adjuvant properties of replication competent contaminating baculovirus. These data demonstrate robust vaccine protection of Gag-based, influenza-pseudotyped VLPs carrying a variety of influenza antigens and suggest applicability toward a number of additional respiratory viruses.


Subject(s)
Gene Products, gag/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Virion/immunology , Animals , Baculoviridae/immunology , Cells, Cultured , Female , Ferrets/immunology , Gene Products, gag/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/isolation & purification , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Male , Mice , Mice, Inbred BALB C , Neuraminidase/immunology , Orthomyxoviridae Infections/immunology , Time Factors
20.
PLoS Pathog ; 4(5): e1000055, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18451982

ABSTRACT

Effective immunotherapies for HIV are needed. Drug therapies are life-long with significant toxicities. Dendritic-cell based immunotherapy approaches are promising but impractical for widespread use. A simple immunotherapy, reinfusing fresh autologous blood cells exposed to overlapping SIV peptides for 1 hour ex vivo, was assessed for the control of SIV(mac251) replication in 36 pigtail macaques. An initial set of four immunizations was administered under antiretroviral cover and a booster set of three immunizations administered 6 months later. Vaccinated animals were randomized to receive Gag peptides alone or peptides spanning all nine SIV proteins. High-level, SIV-specific CD4 and CD8 T-cell immunity was induced following immunization, both during antiretroviral cover and without. Virus levels were durably approximately 10-fold lower for 1 year in immunized animals compared to controls, and a significant delay in AIDS-related mortality resulted. Broader immunity resulted following immunizations with peptides spanning all nine SIV proteins, but the responses to Gag were weaker in comparison to animals only immunized with Gag. No difference in viral outcome occurred in animals immunized with all SIV proteins compared to animals immunized against Gag alone. Peptide-pulsed blood cells are an immunogenic and effective immunotherapy in SIV-infected macaques. Our results suggest Gag alone is an effective antigen for T-cell immunotherapy. Fresh blood cells pulsed with overlapping Gag peptides is proceeding into trials in HIV-infected humans.


Subject(s)
Gene Products, gag/administration & dosage , Immunotherapy/methods , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Viremia/therapy , Animals , Blood Component Transfusion , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Gene Products, gag/immunology , Longevity , Macaca nemestrina , Male , Simian Acquired Immunodeficiency Syndrome/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...