Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
J Virol ; 91(23)2017 12 01.
Article in English | MEDLINE | ID: mdl-28904195

ABSTRACT

An effective AIDS vaccine should elicit strong humoral and cellular immune responses while maintaining low levels of CD4+ T-cell activation to avoid the generation of target cells for viral infection. The present study investigated two prime-boost regimens, both starting vaccination with single-cycle immunodeficiency virus, followed by two mucosal boosts with either recombinant adenovirus (rAd) or fowlpox virus (rFWPV) expressing SIVmac239 or SIVmac251 gag/pol and env genes, respectively. Finally, vectors were switched and systemically administered to the reciprocal group of animals. Only mucosal rFWPV immunizations followed by systemic rAd boost significantly protected animals against a repeated low-dose intrarectal challenge with pathogenic SIVmac251, resulting in a vaccine efficacy (i.e., risk reduction per exposure) of 68%. Delayed viral acquisition was associated with higher levels of activated CD8+ T cells and Gag-specific gamma interferon (IFN-γ)-secreting CD8+ cells, low virus-specific CD4+ T-cell responses, and low Env antibody titers. In contrast, the systemic rFWPV boost induced strong virus-specific CD4+ T-cell activity. rAd and rFWPV also induced differential patterns of the innate immune responses, thereby possibly shaping the specific immunity. Plasma CXCL10 levels after final immunization correlated directly with virus-specific CD4+ T-cell responses and inversely with the number of exposures to infection. Also, the percentage of activated CD69+ CD8+ T cells correlated with the number of exposures to infection. Differential stimulation of the immune response likely provided the basis for the diverging levels of protection afforded by the vaccine regimen.IMPORTANCE A failed phase II AIDS vaccine trial led to the hypothesis that CD4+ T-cell activation can abrogate any potentially protective effects delivered by vaccination or promote acquisition of the virus because CD4+ T helper cells, required for an effective immune response, also represent the target cells for viral infection. We compared two vaccination protocols that elicited similar levels of Gag-specific immune responses in rhesus macaques. Only the animal group that had a low level of virus-specific CD4+ T cells in combination with high levels of activated CD8+ T cells was significantly protected from infection. Notably, protection was achieved despite the lack of appreciable Env antibody titers. Moreover, we show that both the vector and the route of immunization affected the level of CD4+ T-cell responses. Thus, mucosal immunization with FWPV-based vaccines should be considered a potent prime in prime-boost vaccination protocols.


Subject(s)
Fusion Proteins, gag-pol/genetics , Gene Products, env/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL10/blood , Fowlpox virus , Fusion Proteins, gag-pol/immunology , Gene Products, env/immunology , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Immunity, Cellular , Immunity, Humoral , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Vaccination
2.
Clin Vaccine Immunol ; 23(3): 204-12, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26718849

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) acquisition occurs predominantly through mucosal transmission. We hypothesized that greater mucosal immune responses and protective efficacy against mucosal HIV-1 infection may be achieved by prime-boost immunization at mucosal sites. We used a macaque model to determine the safety, immunogenicity, and protective efficacy of orally delivered, replication-competent but attenuated recombinant vaccinia viruses expressing full-length HIV-1 SF162 envelope (Env) or simian immunodeficiency virus (SIV) Gag-Pol proteins. We examined the dose and route that are suitable for oral immunization with recombinant vaccinia viruses. We showed that sublingual inoculation of two vaccinia virus-naive pigtailed macaques with 5 × 10(8) PFU of recombinant vaccinia viruses was safe. However, sublingual inoculation with a higher dose or tonsillar inoculation resulted in secondary oral lesions, indicating the need to optimize the dose and route for oral immunization with replication-competent vaccinia virus vectors. Oral priming alone elicited antibody responses to vaccinia virus and to the SF162 Env protein. Intramuscular immunization with the SF162 gp120 protein at either 20 or 21 weeks postpriming resulted in a significant boost in antibody responses in both systemic and mucosal compartments. Furthermore, we showed that immune responses induced by recombinant vaccinia virus priming and intramuscular protein boosting provided protection against intrarectal challenge with the simian-human immunodeficiency virus SHIV-SF162-P4.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/blood , HIV-1/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , AIDS Vaccines/adverse effects , Animals , Cell Line , Chlorocebus aethiops , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp160/immunology , Immunity, Mucosal/immunology , Immunoglobulin A/blood , Immunoglobulin G/blood , Macaca mulatta/immunology , Vaccination , Vaccines, Synthetic/adverse effects , Vaccinia virus/genetics
3.
PLoS One ; 6(7): e22010, 2011.
Article in English | MEDLINE | ID: mdl-21799765

ABSTRACT

A safe, efficacious vaccine is required to stop the AIDS pandemic. Disappointing results from the STEP trial implied a need to include humoral anti-HIV-1 responses, a notion supported by RV144 trial data even though correlates of protection are unknown. We vaccinated rhesus macaques with recombinant simian immunodeficiency virus (SIV) Gag-Pol particles, HIV-1 Tat and trimeric clade C (HIV-C) gp160, which induced cross-neutralizing antibodies (nAbs) and robust cellular immune responses. After five low-dose mucosal challenges with a simian-human immunodeficiency virus (SHIV) that encoded a heterologous R5 HIV-C envelope (22.1% divergence from the gp160 immunogen), 94% of controls became viremic, whereas one third of vaccinees remained virus-free. Upon high-dose SHIV rechallenge, all controls became infected, whereas some vaccinees remained aviremic. Peak viremia was inversely correlated with both cellular immunity (p<0.001) and cross-nAb titers (p<0.001). These data simultaneously linked cellular as well as humoral immune responses with the degree of protection for the first time.


Subject(s)
HIV Infections/prevention & control , HIV-1/immunology , Simian Immunodeficiency Virus/immunology , Vaccination/methods , Viral Vaccines/immunology , Animals , Fusion Proteins, gag-pol/immunology , HIV Envelope Protein gp160/immunology , HIV Infections/immunology , Humans , Infant , Macaca mulatta , Vaccines, Synthetic/immunology , Viremia/prevention & control , tat Gene Products, Human Immunodeficiency Virus/immunology
4.
Eur J Immunol ; 39(7): 1831-40, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19585509

ABSTRACT

Novel candidate HIV-1 vaccines have been constructed, which are tailor-designed for HLA-B*5101(+) patients infected with HIV-1 clade B. These vaccines employ novel immunogen HIVB-B*5101 derived from consensus HIV-1 clade B Gag p17 and p24 regions coupled to two Pol-derived B*5101-restricted epitopes, which are together with a third B*5101 epitope in Gag dominant in HIV-1-infected long-term non-progressing patients. Both plasmid DNA and modified vaccinia virus Ankara (MVA) vectors supported high expression levels of the HIVB-B*5101 immunogen in cultured cells. Heterologous DNA prime-recombinant MVA boost regimen induced efficiently HIV-1-specific CD8(+) T-cell responses in BALB/c mice. These vaccine-elicited T cells were multifunctional, killed efficiently target cells in vivo, and protected mice against challenge with ecotropic HIV-1/NL4-3 and ecotropic HIV-1/NDK chimaeric viruses with HIV-1 clade B or D backbones, respectively, and ecotropic murine leukemia virus gp80 envelope, and therefore did so in the absence of anti-HIV-1 gp120 antibodies. These results support further development of HIVB-B*5101 vaccines in combined heterologous-modality regimens. The use of allele-specific vaccines in humans is discussed in the context of other developments in the HIV-1 field.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV-1/immunology , HLA-B Antigens/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Amino Acid Sequence , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , DNA, Recombinant/genetics , Epitopes/genetics , Epitopes/immunology , Female , Flow Cytometry , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , HIV Antigens/genetics , HIV Antigens/immunology , HIV Core Protein p24/genetics , HIV Core Protein p24/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/genetics , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Tumor Necrosis Factor-alpha/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
5.
Clin Infect Dis ; 46(11): 1769-81, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18433307

ABSTRACT

BACKGROUND: The safety and immunogenicity of the MRK adenovirus type 5 human immunodeficiency virus type 1 clade B gag/pol/nef vaccine, a replication-incompetent adenovirus type 5-vectored vaccine designed to elicit cell-mediated immunity against conserved human immunodeficiency virus proteins, was assessed in a phase 1 trial. METHODS: Healthy adults not infected with human immunodeficiency virus were enrolled in a multicenter, dose-escalating, blind, placebo-controlled study to evaluate a 3-dose homologous prime-boost regimen of the trivalent MRK adenovirus type 5 human immunodeficiency virus type 1 vaccine containing from 3 x 10(6) to 1 x 10(11) viral particles per 1-mL dose administered on day 1, during week 4 and during week 26. Adverse events were recorded for 29 days after each intradeltoid injection. The primary immunogenicity end point was the proportion of study participants with a positive unfractionated Gag-, Pol-, or Nef-specific interferon-gamma enzyme-linked immunosorbent spot response measured 4 weeks after administration of the last dose. RESULTS: Of 259 randomized individuals, 257 (99%) received > or = 1 dose of vaccine or placebo and were included in the safety analyses. Enzyme-linked immunosorbent spot results were available for 217 study participants (84%) at week 30. No serious vaccine-related adverse events occurred. No study participant discontinued participation because of vaccine-related adverse events. The frequency of injection-site reactions was dose dependent. Vaccine doses of > or = 3 x 10(9) viral particles elicited positive enzyme-linked immunosorbent spot responses to > or = 1 vaccine component in > 60% of recipients. High baseline antibody titers against adenovirus type 5 diminished enzyme-linked immunosorbent spot responses at all doses except the 3 x 10(10) viral particle dose. CONCLUSIONS: The vaccine was generally well tolerated and induced cell-mediated immune responses against human immunodeficiency virus type 1 peptides in most healthy adults. Despite these findings, vaccination in a proof-of-concept trial with use of this vaccine was discontinued because of lack of efficacy.


Subject(s)
AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , HIV Infections/prevention & control , HIV-1/immunology , AIDS Vaccines/administration & dosage , Adenoviridae , Adult , Female , Fusion Proteins, gag-pol/immunology , Genes, gag , Genes, pol , HIV Antibodies/biosynthesis , HIV Infections/immunology , HIV-1/genetics , Humans , Male , Safety , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology
6.
Eur J Immunol ; 38(5): 1310-20, 2008 May.
Article in English | MEDLINE | ID: mdl-18412164

ABSTRACT

The development of an HIV vaccine that induces broad and potent immunity is critically needed. Viruses, including lentiviruses, have been used as vectors for ex vivo transduction of antigens into dendritic cells (DC). We hypothesized that DC transduced with a vector that allows selective infection of DC could induce potent immunity by continually priming DC. A lentiviral vector encoding HIV gag-pol without env would form viral cores in transduced DC, but would release non-infectious particles by budding into endosomes and releasing apoptotic bodies or exosomes containing viral cores. DC function by endocytosing DC-derived apoptotic bodies, and they are specialized in their ability to move endocytic contents into the cytoplasm. We postulated that endocytosis of vector cores could lead to transduction of a second round of DC. In this report, we demonstrate accumulation of viral cores inside transduced DC and show second-round transduction of immature DC that endocytose transduced DC in vitro. The effectiveness of immunization of mice with transduced DC to induce specific lymphocyte activation was assessed. Mice developed antigen-specific T cell responses and specific antibodies after immunization. Transduction of DC with a replication-competent but conditionally infectious lentivirus could be a novel vaccine strategy for HIV.


Subject(s)
AIDS Vaccines/immunology , B-Lymphocytes/immunology , Lentivirus/immunology , T-Lymphocytes/immunology , Vaccination/methods , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Antibody Formation/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Dendritic Cells/virology , Female , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Genetic Vectors/genetics , HIV/immunology , HIV Core Protein p24/immunology , HIV Core Protein p24/metabolism , Interferon-gamma/metabolism , Lentivirus/genetics , Mice , Mice, Inbred BALB C , Microscopy, Electron , Nucleocapsid/biosynthesis , Nucleocapsid/ultrastructure , Protein Precursors/analysis , Protein Precursors/metabolism , Transduction, Genetic , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
7.
Virology ; 374(2): 528-34, 2008 May 10.
Article in English | MEDLINE | ID: mdl-18272194

ABSTRACT

Persistent gag-specific T cell immunity would be a useful component of an effective HIV vaccine. The Flavivirus Kunjin replicon was previously engineered to persistently express HIV gag and was shown to induce protective responses in mice. We evaluated Kunjin replicon virus-like-particles expressing SIVgag-pol in pigtail macaques. Kunjin-specific antibodies were induced, but no SIV-specific T cell immunity were detected. Following SIVmac251 challenge, there was no difference in SIV viremia or retention of CD4 T cells between Kunjin-SIVgag-pol vaccine immunized animals and controls. An amnestic SIV gag-specific CD8 T cell response associated with control of viremia was observed in 1 of 6 immunized animals. Refinements of this vector system and optimization of the immunization doses, routes, and schedules are required prior to clinical trials.


Subject(s)
Replicon , SAIDS Vaccines , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vaccines, Synthetic/immunology , West Nile virus , AIDS Vaccines , Animals , Antibodies, Viral/blood , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Fusion Proteins, gag-pol/metabolism , Genetic Engineering , HIV-1/genetics , HIV-1/immunology , HIV-1/metabolism , Lymphocyte Activation , Macaca nemestrina , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Simian Immunodeficiency Virus/pathogenicity , T-Lymphocytes/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/metabolism , West Nile virus/genetics , West Nile virus/immunology , West Nile virus/metabolism
8.
Vaccine ; 25(35): 6474-81, 2007 Aug 29.
Article in English | MEDLINE | ID: mdl-17688978

ABSTRACT

We reported previously the absence of systemic infection in a subset of macaques vaccinated with an HIV-1 DNA/MVA vaccine after 18 or more rectal challenges with low (physiologically relevant) doses of SHIV162P3. To further study the apparent protection, we looked for sequestered virus in gut tissues, lymph nodes, spleen, and testes obtained at necropsy using virus co-culture and nested PCR for SIV Gag, Pol and LTR. There was no evidence of sequestered virus in tissues obtained from the four protected macaques. In contrast, at least one tissue from each of 11 infected animals scored positive by one of these sensitive procedures. Activated PBMC from the protected macaques were not inherently resistant to in vitro infection by the challenge virus. These findings demonstrate that some vaccinated macaques appeared to be free from the challenge virus. Therefore, such T cell-based vaccines may provide some protection when challenge virus doses approach physiological equivalencies.


Subject(s)
AIDS Vaccines/therapeutic use , Digestive System/virology , Lymph Nodes/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fusion Proteins, gag-pol/immunology , HIV Core Protein p24/immunology , HIV-1/immunology , Immunity, Innate/immunology , Lymphocytes/virology , Lymphoid Tissue/virology , Macaca mulatta , Monocytes/virology , Rectum/virology , Reverse Transcriptase Polymerase Chain Reaction , Vaccines, DNA/immunology
9.
J Med Primatol ; 36(1): 2-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17359459

ABSTRACT

BACKGROUND: The long-term benefits of highly active antiretroviral therapy in HIV-infected patients are limited by emergence of drug-resistant variants and side effects. Therefore, we studied the concept of therapeutic immunization in 18 rhesus monkeys infected with a highly pathogenic simian immunodeficiency virus (SIV) swarm. METHODS: Monkeys were treated with the reverse transcriptase inhibitor (R)-9-(2-phosphonylmethoxypropyl)adenine (PMPA) for 19 weeks starting 10 days after infection. After suppression of viremia, one group of monkeys was immunized with recombinant modified vaccinia virus Ankara (MVA) vectors expressing gag-pol and env. A second group received MVA vectors expressing the regulatory genes tat, rev and nef, while a third group was not immunized. RESULTS: Immunization with gag-pol and env expressing MVA enhanced SIV antibody titers. Following discontinuation of PMPA treatment, a rebound in viral load was observed. However, in three of six monkeys immunized with MVA gag-pol and MVA env, and two of six monkeys immunized MVA expressing regulatory genes set point RNA levels were below or close to a threshold level of 10(4) RNA copies/ml, while only one of six unvaccinated monkeys maintained such low RNA levels. CONCLUSIONS: Although a subset of animals seem to benefit from therapeutic immunization with MVA vectors, the difference in set point RNA levels between the groups did not reach statistical significance.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Macaca mulatta/immunology , Macaca mulatta/virology , SAIDS Vaccines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Adenine/analogs & derivatives , Adenine/therapeutic use , Animals , Antibodies, Viral/blood , Fusion Proteins, gag-pol/immunology , Genetic Vectors , Organophosphonates/therapeutic use , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Tenofovir , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Vaccinia virus/genetics
10.
Vaccine ; 25(15): 2863-85, 2007 Apr 12.
Article in English | MEDLINE | ID: mdl-17113200

ABSTRACT

In this investigation we have generated and defined the immunogenicity of two novel HIV/AIDS vaccine candidates based on the highly attenuated vaccinia virus strains, MVA and NYVAC, efficiently expressing in the same locus (TK) and under the same viral promoter the codon optimized HIV-1 genes encoding gp120 and Gag-Pol-Nef antigens of clade B (referred as MVA-B and NYVAC-B). In infected human HeLa cells, gp120 is released from cells and GPN is produced as a polyprotein; NYVAC-B induces severe apoptosis but not MVA-B. The two poxvirus vectors showed genetic stability of the inserts. In BALB/c and in transgenic HHD mice for human HLA-A2 class I, both vectors are efficient immunogens and induced broad cellular immune responses against peptides represented in the four HIV-1 antigens. Some differences were observed in the magnitude and breadth of the immune response in the mouse models. In DNA prime/poxvirus boost protocols, the strongest immune response, as measured by fresh IFN-gamma and IL-2 ELISPOT, was obtained in BALB/c mice boosted with NYVAC-B, while in HHD mice there were no differences between the poxvirus vectors. When the prime/boost was performed with homologous or with combination of poxvirus vectors, the protocols MVA-B/MVA-B and NYVAC-B/NYVAC-B, or the combination NYVAC-B/MVA-B gave the most consistent broader immune response in both mouse models, although the magnitude of the overall response was higher for the DNA-B/poxvirus-B regime. All of the immunization protocols induced some humoral response against the gp160 protein from HIV-1 clone LAV. Our findings indicate that MVA-B and NYVAC-B meet the criteria to be potentially useful vaccine candidates against HIV/AIDS.


Subject(s)
AIDS Vaccines/immunology , Antigens, Viral/immunology , HIV Envelope Protein gp120/immunology , Viral Vaccines/immunology , AIDS Vaccines/genetics , Animals , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Apoptosis/immunology , Base Sequence , Chick Embryo , Fusion Proteins, gag-pol/biosynthesis , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Gene Products, nef/biosynthesis , Gene Products, nef/genetics , Gene Products, nef/immunology , Genomic Instability , HIV Envelope Protein gp120/biosynthesis , HIV Envelope Protein gp120/genetics , HLA-A2 Antigen/immunology , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Polymerase Chain Reaction/methods , Poxviridae/genetics , Poxviridae/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/genetics
11.
J Infect Dis ; 194(12): 1638-49, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17109335

ABSTRACT

BACKGROUND: The development of an effective human immunodeficiency virus (HIV) vaccine is a high global priority. Here, we report the safety, tolerability, and immunogenicity of a replication-defective recombinant adenovirus serotype 5 (rAd5) vector HIV-1 candidate vaccine. METHODS: The vaccine is a mixture of 4 rAd5 vectors that express HIV-1 subtype B Gag-Pol fusion protein and envelope (Env) from subtypes A, B, and C. Healthy, uninfected adults were randomized to receive 1 intramuscular injection of placebo (n=6) or vaccine at dose levels of 10(9) (n=10), 10(10) (n=10), or 10(11) (n=10) particle units and were followed for 24 weeks to assess immunogenicity and safety. RESULTS: The vaccine was well tolerated but was associated with more reactogenicity at the highest dose. At week 4, vaccine antigen-specific T cell responses were detected in 28 (93.3%) and 18 (60%) of 30 vaccine recipients for CD4(+) and CD8(+) T cells, respectively, by intracellular cytokine staining assay and in 22 (73%) of 30 vaccine recipients by enzyme-linked immunospot assay. Env-specific antibody responses were detected in 15 (50%) of 30 vaccine recipients by enzyme-linked immunosorbant assay and in 28 (93.3%) of 30 vaccine recipients by immunoprecipitation followed by Western blotting. No neutralizing antibody was detected. CONCLUSIONS: A single injection induced HIV-1 antigen-specific CD4(+) T cell, CD8(+) T cell, and antibody responses in the majority of vaccine recipients. This multiclade rAd5 HIV-1 vaccine is now being evaluated in combination with a multiclade HIV-1 DNA plasmid vaccine.


Subject(s)
AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Nausea/etiology , Vaccination , AIDS Vaccines/administration & dosage , Adenoviruses, Human/genetics , Adolescent , Adult , Antibody Specificity , Blotting, Western , Cytokines/analysis , Cytokines/biosynthesis , Dose-Response Relationship, Immunologic , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fusion Proteins, gag-pol/immunology , Gene Products, env/immunology , Genetic Vectors , Humans , Injections, Intramuscular , Male , Recombination, Genetic , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , env Gene Products, Human Immunodeficiency Virus
12.
J Infect Dis ; 194(12): 1650-60, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17109336

ABSTRACT

BACKGROUND: Gene-based vaccine delivery is an important strategy in the development of a preventive vaccine for acquired immunodeficiency syndrome (AIDS). Vaccine Research Center (VRC) 004 is the first phase 1 dose-escalation study of a multiclade HIV-1 DNA vaccine. METHODS: VRC-HIVDNA009-00-VP is a 4-plasmid mixture encoding subtype B Gag-Pol-Nef fusion protein and modified envelope (Env) constructs from subtypes A, B, and C. Fifty healthy, uninfected adults were randomized to receive either placebo (n=10) or study vaccine at 2 mg (n=5), 4 mg (n=20), or 8 mg (n=15) by needle-free intramuscular injection. Humoral responses (measured by enzyme-linked immunosorbant assay, Western blotting, and neutralization assay) and T cell responses (measured by enzyme-linked immunospot assay and intracellular cytokine staining after stimulation with antigen-specific peptide pools) were measured. RESULTS: The vaccine was well tolerated and induced cellular and humoral responses. The maximal CD4(+) and CD8(+) T cell responses occurred after 3 injections and were in response to Env peptide pools. The pattern of cytokine expression by vaccine-induced HIV-specific T cells evolved over time, with a diminished frequency of interferon- gamma -producing T cells and an increased frequency of interleukin-2-producing T cells at 1 year. CONCLUSIONS: DNA vaccination induced antibody to and T cell responses against 3 major HIV-1 subtypes and will be further evaluated as a potential component of a preventive AIDS vaccine regimen.


Subject(s)
AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Genetic Vectors , HIV Infections/immunology , HIV-1/immunology , Immunization Schedule , Neutralization Tests , Plasmids , Vaccination , AIDS Vaccines/administration & dosage , Adolescent , Adult , Antibodies, Viral/blood , Antibody Specificity , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/analysis , Cytokines/biosynthesis , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Gene Products, nef/genetics , Gene Products, nef/immunology , HIV Infections/blood , Humans , Injections, Intramuscular , Male , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , nef Gene Products, Human Immunodeficiency Virus
13.
Clin Vaccine Immunol ; 13(7): 747-57, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16829611

ABSTRACT

The FP9 strain of F has been described as a more immunogenic recombinant vaccine vector than the Webster FPV-M (FPW) strain (R. J. Anderson et al., J. Immunol. 172:3094-3100, 2004). This study expands the comparison to include two separate recombinant antigens and multiple, rather than single, independent viral clones derived from the two strains. Dual-poxvirus heterologous prime-boost vaccination regimens using individual clones of recombinant FP9 or FPW in combination with recombinant modified V Ankara expressing the same antigen were evaluated for their ability to elicit T-cell responses against recombinant antigens from Plasmodium berghei (circumsporozoite protein) or human immunodeficiency virus type 1 (a Gag-Pol-Nef fusion protein). Gamma interferon enzyme-linked immunospot assay and fluorescence-activated cell sorting assays of the responses to specific epitopes confirmed the approximately twofold-greater cellular immunogenicity of FP9 compared to FPW, when given as the priming or boosting immunization. Equality of transgene expression in mouse cells infected with the two strains in vitro was verified by Western blotting. Directed partial sequence analysis and PCR analysis of FPW and comparison to available whole-genome sequences revealed that many loci that are mutated in the highly attenuated and culture-adapted FP9 strain are wild type in FPW, including the seven multikilobase deletions. These "passage-specific" alterations are hypothesized to be involved in determining the immunogenicity of fowlpox virus as a recombinant vaccine vector.


Subject(s)
Fowlpox virus/classification , Fowlpox virus/immunology , Genetic Vectors/immunology , HIV-1/genetics , Plasmodium berghei/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Female , Fowlpox virus/genetics , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Gene Products, nef/genetics , Gene Products, nef/immunology , Gene Products, nef/metabolism , HIV-1/immunology , Humans , Immunization, Secondary , Interferon-gamma/metabolism , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , Polyproteins/genetics , Polyproteins/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccination , nef Gene Products, Human Immunodeficiency Virus
14.
Vaccine ; 24(21): 4503-9, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16181711

ABSTRACT

A plasmid DNA vaccine containing a fusion gene consisting of an HIV-1 subtype C gag and a modified subtype C pol was compared to a mixture of gag plus pol or gag plus HIV env plasmids. Plasmid DNA was delivered by intramuscular injection followed by electroporation in vivo. Two vaccinations were sufficient to induce high levels of Gag- and Pol-specific CD4 and CD8 T cells in peripheral blood. The gag-pol fusion plasmid was as immunogenic as the plasmid mixtures. Thus, DNA vaccination by intramuscular electroporation was an effective means for inducing high levels of Gag- and Pol-specific T cells, and a single gag-pol fusion DNA vaccine was sufficient for eliciting immune responses against both antigens.


Subject(s)
AIDS Vaccines/immunology , Electroporation/methods , Fusion Proteins, gag-pol/immunology , HIV-1/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Macaca mulatta , Vaccines, DNA/administration & dosage
15.
J Immunol ; 173(7): 4387-93, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383568

ABSTRACT

Several gene-based vaccine approaches are being tested to drive multivalent cellular immune responses to control HIV-1 viral variants. To compare the utility of these approaches, HLA-A*0201 transgenic mice were genetically immunized with plasmids encoding wild-type (wt) gag-pol, codon-optimized (CO) gag-pol, and an expression library immunization (ELI) vaccine genetically re-engineered to express non-CO fragments of gag and pol fused to ubiquitin for proteasome targeting. Equimolar delivery of each vaccine into HLA-A*0201 transgenic mice generated CD8 T cell responses, with the ELI vaccine producing up to 10-fold higher responses than the wt or CO gag-pol plasmids against cognate and mutant epitopes. All three vaccines generated multivalent CD8 responses against varying numbers of epitopes after priming. However, when the animals were immunized again, the wt and CO gag-pol vaccines boosted only the responses against a subset of epitopes and attenuated the responses against all other Ags including epitopes from clade and drug-resistant viral variants. In contrast, the ELI vaccine boosted CD8 responses against all of the gag-pol Ags and against mutant epitopes from clade and drug-resistant variants. These data suggest that HIV-1 vaccines expressing structurally intact gag and pol proteins drive immunofocused CD8 responses that reduce the repertoire of T cell responses. In contrast, the genetically re-engineered ELI vaccine appears to better maintain the multivalent CD8 responses that may be required to control HIV-1 viral variants.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fusion Proteins, gag-pol/immunology , Gene Library , HIV-1/immunology , Lymphocyte Activation , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , CD8-Positive T-Lymphocytes/virology , Cell Line , Dose-Response Relationship, Immunologic , Drug Resistance, Viral/genetics , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Fusion Proteins, gag-pol/administration & dosage , Fusion Proteins, gag-pol/genetics , H-2 Antigens/genetics , HIV Protease/genetics , HIV-1/enzymology , HIV-1/genetics , HLA-A Antigens/genetics , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Immunization, Secondary , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Interferon-gamma/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C3H , Mice, Transgenic , Peptide Library , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
16.
AIDS Res Hum Retroviruses ; 20(8): 846-59, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15320989

ABSTRACT

We explored the use of a simian-human immunodeficiency virus (SHIV) DNA vaccine as an effective mucosal priming agent to stimulate a protective immune response for AIDS prevention. Rhesus macaques were vaccinated rectally with a DNA construct producing replication-defective SHIV particles, and boosted with either the same DNA construct or recombinant modified vaccinia virus Ankara (MVA) expressing SIV Gag, SIV Pol, and HIV Env (MVA-SHIV). Virus-specific mucosal and systemic humoral and cell-mediated immune responses could be stimulated by this approach but were present inconsistently among the vaccinated animals. Rectal vaccination with either SHIV DNA alone or SHIV DNA followed by MVA-SHIV induced SIV Gag/Pol- or HIV gp120-specific IgA in rectal secretions of four of seven animals. However, the gp120-specific rectal IgA antibody responses were not durable and had become undetectable in all but one animal shortly before rectal challenge with pathogenic SHIV 89.6P. Only the macaques primed with SHIV DNA and boosted with MVA-SHIV demonstrated SHIV-specific IgG in plasma. In addition, these animals developed more consistent antiviral cell-mediated responses and had better preservation of CD4 T cells following challenge with SHIV 89.6P. Our study demonstrates the utility of a rectal DNA/MVA vaccination protocol for the induction of diverse responses in different immunological compartments. In addition, the immunity achieved with this mucosal vaccination regimen is sufficient to delay progression to AIDS.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , AIDS Vaccines/genetics , Administration, Rectal , Animals , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Immunity, Mucosal , Macaca mulatta , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccinia virus/genetics , Vaccinia virus/immunology , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology
17.
AIDS Res Hum Retroviruses ; 20(6): 642-4, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15242541

ABSTRACT

A phase I/II prime-boost vaccine trial in HIV-1-seronegative adults was conducted in Thailand using ALVAC-HIV (vCP1521) as a prime, boosting with either oligomeric gp160 TH023/LAI or Chiron HIV Thai subtype E (CM235) plus U. S. subtype B (SF2) gp120. Cytotoxic T lymphocyte (CTL) assays were conducted at one of the vaccine trial sites (Siriraj Hospital) at a single time point following the completion of immunization demonstrated that 8 of 50 (16%) vaccine recipients showed HIV-specific CTL by standard chromium release assay (CRA) after in vitro stimulation (IVS) for 2 weeks. Five additional vaccinees (13/50 = 26%) showed CTL responses after IVS for up to 4 weeks. Moreover, one volunteer with a positive CTL response to a single HIV antigen at Day 14 demonstrated a response to an additional HIV-1 antigen(s) after the longer IVS period. CTL activity was CD8+ restricted. Despite extension of the IVS up to 4 weeks, no CTL responses were detected in placebo recipients. These results imply that extension of the IVS period may increase the sensitivity of the CRA when measuring HIV-specific CTL in ALVAC-HIV prime-boost recipients without compromising specificity.


Subject(s)
AIDS Vaccines/immunology , Cytotoxicity, Immunologic , HIV Infections/prevention & control , HIV-1/immunology , T-Lymphocytes, Cytotoxic/immunology , AIDS Vaccines/administration & dosage , Chromium Radioisotopes , Cytotoxicity Tests, Immunologic , Fusion Proteins, gag-pol/immunology , Gene Products, env/immunology , Humans , Immunization, Secondary , Lymphocyte Activation , Sensitivity and Specificity , Thailand , Time Factors , Vaccination
18.
J Virol ; 78(14): 7490-7, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15220422

ABSTRACT

Because a strategy to elicit broadly neutralizing anti-human immunodeficiency virus type 1 (HIV-1) antibodies has not yet been found, the role of an Env immunogen in HIV-1 vaccine candidates remains undefined. We sought to determine whether an HIV-1 Env immunogen genetically disparate from the Env of the challenge virus can contribute to protective immunity. We vaccinated Indian-origin rhesus monkeys with Gag-Pol-Nef immunogens, alone or in combination with Env immunogens that were either matched or mismatched with the challenge virus. These animals were then challenged with a pathogenic simian-human immunodeficiency virus. The vaccine regimen included a plasmid DNA prime and replication-defective adenoviral vector boost. Vaccine regimens that included the matched or mismatched Env immunogens conferred better protection against CD4(+) T-lymphocyte loss than that seen with comparable regimens that did not include Env immunogens. This increment in protective immunity was associated with anamnestic Env-specific cellular immunity that developed in the early days following viral challenge. These data suggest that T-lymphocyte immunity to Env can broaden the protective cellular immune response to HIV despite significant sequence diversity of the strains of the Env immunogens and can contribute to immune protection in this AIDS vaccine model.


Subject(s)
AIDS Vaccines/immunology , Gene Products, env/immunology , HIV Infections/prevention & control , Recombinant Proteins/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Adenoviridae/genetics , Adenoviridae/immunology , Animals , CD4 Lymphocyte Count , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Gene Products, env/genetics , Gene Products, nef/genetics , Gene Products, nef/immunology , HIV-1/immunology , Humans , Macaca mulatta , RNA, Viral/blood , SAIDS Vaccines/administration & dosage , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , nef Gene Products, Human Immunodeficiency Virus
19.
AIDS Res Hum Retroviruses ; 20(4): 425-34, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15157361

ABSTRACT

We utilized SIV(mne) infection of Macaca fascicularis to assess the efficacy of DNA vaccination alone, and as a priming agent in combination with subunit protein boosts. All SIV(mne) structural and regulatory genes were expressed using the human cytomegalovirus Immediate Early-1 promoter in plasmids that directed the formation of virus-like particles in vitro. Macaques (n = 4) were immunized intradermally and intramuscularly four times over 36 weeks with 3 mg plasmid DNA. A second group (n = 4) received two DNA priming inoculations followed by two intramuscular boosts consisting of 250 microg recombinant Env gp160 and 250 microg recombinant Gag-Pol particles in MF-59 adjuvant. These regimens elicited modest cellular immunity prior to challenge. Humoral immune responses to Env gp160 were elicited and sustained by both vaccine protocols, and as expected antibody titers were higher in the protein subunit-boosted animals. Neutralizing antibodies prior to challenge were measurable in two of four subunit-boosted macaques. The two vaccine regimens elicited comparable helper T cell responses at the time of challenge. Vaccinees and mock-immunized controls (n = 4) were challenged intrarectally at week 38 with uncloned SIV(mne). Following challenge all macaques became infected, but both vaccine regimens resulted in reduced peak virus loads (p = 0.07) and significantly improved maintenance of peripheral CD4(+) T cell counts postchallenge (p = 0.007, DNA alone and p = 0.01, all vaccinees). There was no significant difference between the two vaccine groups in levels of plasma viremia or maintenance of CD4(+) T cell counts postchallenge.


Subject(s)
SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , CD4 Lymphocyte Count , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Gene Products, env/immunology , HIV Envelope Protein gp160/genetics , HIV Envelope Protein gp160/immunology , Immunity, Cellular , Macaca fascicularis , Neutralization Tests , Plasmids , Proviruses/genetics , Proviruses/isolation & purification , RNA, Viral/blood , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Vaccines, DNA/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Load
20.
AIDS Res Hum Retroviruses ; 19(10): 891-900, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14585221

ABSTRACT

Envelope protein immunogens may improve DNA or live-vectored HIV vaccines by complementing antiviral cellular responses with Env antibodies. We tested this concept by administering two immunizations of alum-adjuvanted HIV-1 89.6 gp120 to macaques being primed at weeks 0 and 8 with SHIV 89.6 Gag-Pol-Env DNA and boosted at week 24 with SHIV-89.6 Gag-Pol-Env recombinant modified vaccinia Ankara (MVA). Three hundred micrograms of gp120 was delivered with the second DNA prime and the MVA booster. Eight months after vaccination, all animals were challenged intrarectally with the related, yet serologically distinct, SHIV-89.6P. The gp120 immunizations raised binding, but not neutralizing antibody for the challenge virus, and allowed testing of whether gp120 vaccines that fail to raise neutralizing antibody can improve protection. Following the second gp120 immunization, the plus-gp120 group showed >10 times higher levels of binding antibody than the minus-gp120 group. These levels fell and were overall similar in both groups at the time of challenge. Following the second challenge, both groups had similar temporal patterns and heights of binding and neutralizing antibodies. However, the plus-gp120 group had less consistent control of viremia and higher levels of plasma viral RNA for the first year postchallenge. Assays for complement-dependent enhancing antibody revealed a trend toward higher levels of activity in the plus-gp120 group. This trend did not reach significance in our animal groups of 8. We conclude that gp120 inoculations that fail to raise neutralizing antibody do not improve the efficacy of Gag-Pol-Env DNA/MVA vaccines.


Subject(s)
AIDS Vaccines/immunology , Fusion Proteins, gag-pol/immunology , Gene Products, env/immunology , HIV Envelope Protein gp120/immunology , Vaccinia virus/immunology , AIDS Vaccines/administration & dosage , Animals , HIV Envelope Protein gp120/genetics , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Vaccines, DNA/immunology , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...