Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Vaccine ; 26(37): 4819-29, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18657584

ABSTRACT

The effectiveness of plasmid DNA (pDNA) vaccines can be improved by the co-delivery of plasmid-encoded molecular adjuvants. We evaluated pDNAs encoding GM-CSF, Flt-3L, IL-12 alone, or in combination, for their relative ability to serve as adjuvants to augment humoral and cell-mediated immune responses elicited by prototype pDNA vaccines. In Balb/c mice we found that co-administration of plasmid-based murine GM-CSF (pmGM-CSF), murine Flt-3L (pmFlt-3L) or murine IL-12 (pmIL-12) could markedly enhance the cell-mediated immune response elicited by an HIV-1 env pDNA vaccine. Plasmid mGM-CSF also augmented the immune response elicited by DNA vaccines expressing HIV-1 Gag and Nef-Tat-Vif. In addition, the use of pmGM-CSF as a vaccine adjuvant appeared to markedly increase antigen-specific proliferative responses and improved the quality of the resulting T-cell response by increasing the percentage of polyfunctional memory CD8(+) T cells. Co-delivery of pmFlt-3L with pmGM-CSF did not result in a further increase in adjuvant activity. However, the co-administration of pmGM-CSF with pmIL-12 did significantly enhance env-specific proliferative responses and vaccine efficacy in the murine vaccinia virus challenge model relative to mice immunized with the env pDNA vaccine adjuvanted with either pmGM-CSF or pmIL-12 alone. These data support the testing of pmGM-CSF and pmIL-12, used alone or in combination, as plasmid DNA vaccine adjuvants in future macaque challenge studies.


Subject(s)
AIDS Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-12/pharmacology , Membrane Proteins/pharmacology , Vaccines, DNA/immunology , Adjuvants, Immunologic/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , HIV-1/genetics , HIV-1/immunology , Interleukin-12/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Plasmids , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/immunology
2.
Vaccine ; 26(40): 5083-94, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18485543

ABSTRACT

Plasmid DNA (pDNA) vaccines are effective at eliciting immune responses in a wide variety of animal model systems, however, pDNA vaccines have generally been incapable of inducing robust immune responses in clinical trials. Therefore, to identify means to improve pDNA vaccine performance, we compared various post-transcriptional and post-translational genetic modifications for their ability to improve antigen-specific CMI responses. Mice vaccinated using a sub-optimal 100 mcg dose of a pDNA encoding an unmodified primary isolate HIV-1(6101) env gp160 failed to demonstrate measurable env-specific CMI responses. In contrast, significant env-specific CMI responses were seen in mice immunized with pDNA expression vectors encoding env genes modified by RNA optimization or codon optimization. Further modification of the RNA optimized env gp160 gene by the addition of (i) a simian retrovirus type 1 constitutive RNA transport element; (ii) a murine intracisternal A-particle derived RNA transport element; (iii) a tissue plasminogen activator protein signal leader sequences; (iv) a beta-catenin derived ubiquitination target sequence; or (v) a monocyte chemotactic protein-3 derived signal sequence failed to further improve the induction of env-specific CMI responses. Therefore, modification of the env gp160 gene by RNA or codon optimization alone is necessary for high-level rev-independent expression and results in robust env-specific CMI responses in immunized mice. Importantly, further modification(s) of the env gene to alter cellular localization or increase proteolytic processing failed to result in increased env-specific immune responses. These results have important implications for the design and development of an efficacious vaccine for the prevention of HIV-1 infection.


Subject(s)
AIDS Vaccines/immunology , Codon/genetics , HIV Envelope Protein gp160/genetics , HIV Infections/prevention & control , RNA/genetics , T-Lymphocytes/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Female , HIV Antibodies/blood , HIV Envelope Protein gp160/immunology , HIV Envelope Protein gp160/metabolism , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , HIV-1/metabolism , Immunity, Cellular , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Plasmids/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
3.
Vaccine ; 25(26): 4967-82, 2007 Jun 21.
Article in English | MEDLINE | ID: mdl-17335943

ABSTRACT

Plasmid-based IL-12 has been demonstrated to successfully enhance the immunogenicity of DNA vaccines, thus enabling a reduction of the amount of DNA required for immunization. IL-15 is thought to affect the maintenance and enhance effector function of CD8(+) memory T cells. Since the ability to elicit a long-term memory response is a desirable attribute of a prophylactic vaccine, we sought to evaluate the ability of these plasmid-based cytokines to serve as vaccine adjuvants in rhesus macaques. Macaques were immunized with plasmid DNA encoding SIVgag in combination with plasmid IL-12, IL-15, or a combination of IL-12 and IL-15. The plasmid-based cytokines were monitored for their ability to augment SIVgag-specific cellular and humoral immune responses and to alter the clinical outcome following pathogenic SHIV(89.6P) challenge. Macaques receiving SIVgag pDNA in combination with plasmid IL-12 alone, or in combination with plasmid IL-12 and IL-15, demonstrated significantly elevated cell-mediated and humoral immune responses resulting in an improved clinical outcome following virus challenge compared to macaques receiving SIVgag pDNA alone. Macaques receiving SIVgag pDNA in combination with plasmid IL-15 alone demonstrated minor increases in cell-mediated and humoral immune responses, however, the clinical outcome following virus challenge was not improved. These results have important implications for the continued development of plasmid DNA vaccines for the prevention of HIV-1 infection.


Subject(s)
Antibody Formation/genetics , Antibody Formation/immunology , Gene Products, gag/genetics , Gene Products, gag/immunology , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Interleukin-12/genetics , Interleukin-15/genetics , Plasmids/genetics , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Progression , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/immunology , Macaca mulatta , Male , Neutralization Tests , Vaccines, DNA/immunology
4.
Vaccine ; 24(21): 4677-87, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16288822

ABSTRACT

An experimental pDNA vaccine adjuvant expressing IL-12 was evaluated for its ability to augment the humoral and cellular immune responses elicited by a SIVmac239 gag p39 expressing pDNA vaccine. To determine the effect of vaccine dose on the immune response, rhesus macaques were immunized with 1.5 mg or 5.0 mg of SIVmac239 gag pDNA, with or without co-immunization of IL-12 pDNA at 1.5 mg and 5.0 mg, respectively. Serum antibody responses to simian immunodeficiency virus (SIV) gag were increased 10-fold (p=0.044, 0.002) in macaques receiving IL-12 pDNA. Cellular immune responses, monitored by SIV gag-specific IFN-gamma ELISpot assay, were also significantly higher (p=0.007, 0.019) when the pDNA vaccine was co-immunized with IL-12 pDNA at high and low doses. There was no statistical difference between the immune responses elicited by the high and low dose of IL-12 pDNA (p=0.221, 0.917), a finding which could allow a dose reduction of vaccine without the concomitant loss of imunogenicity. Furthermore, analysis of the breadth of the T-cell response during the vaccination schedule, using overlapping peptides to SIV gag, demonstrated a significant correlation (p=0.0002) between the magnitude and breadth of the immune responses in the vaccines. These results have important implications for the continuing development of an effective, safe low dose pDNA vaccine adjuvant suitable for human use.


Subject(s)
Adjuvants, Immunologic/pharmacology , Gene Products, gag/genetics , Interleukin-12/pharmacology , Plasmids , Simian Immunodeficiency Virus/genetics , Vaccines, DNA/administration & dosage , Adjuvants, Immunologic/genetics , Animals , Antibodies, Viral/biosynthesis , Enzyme-Linked Immunosorbent Assay , Immunity, Cellular , Interleukin-12/genetics , Macaca mulatta
5.
AIDS Res Hum Retroviruses ; 20(9): 989-1004, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15585086

ABSTRACT

An experimental AIDS vaccine based on attenuated, recombinant vesicular stomatitis virus (rVSV), when administered by a combination of parenteral and mucosal routes, has proven effective at preventing AIDS in a rhesus macaque model (Rose NF, et al.: Cell 2001;106:539-549). In an effort to determine the optimal route of vaccine administration we evaluated the ability of rVSV-based vaccine vectors expressing HIV-1 Env and SIV Gag proteins, when given either intramuscularly (i.m.) or intranasally (i.n.), to elicit antigen-specific cellular and humoral immune responses, and to protect from a subsequent vaginal challenge with simian-human immunodeficiency virus (SHIV89.6P). Our results demonstrate that macaques vaccinated by the i.n. route developed significantly higher antigen-specific cellular immune responses as determined by MHC class I tetramer staining, IFN-gamma ELISPOT, and cytotoxic T cell assays. However, systemic and mucosal humoral immune responses did not vary significantly with the route of vaccine administration. Given the importance of cell-mediated immune responses in slowing AIDS progression, intranasal delivery of a VSV-based AIDS vaccine may be an optimal as well as practical route for vaccination and should be considered in design of clinical trials.


Subject(s)
AIDS Vaccines/immunology , Gene Products, env/immunology , Gene Products, gag/immunology , Genetic Vectors/immunology , Vaccines, Attenuated/immunology , Vesicular stomatitis Indiana virus/immunology , AIDS Vaccines/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/blood , Female , Gene Products, env/genetics , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Genetic Vectors/administration & dosage , HIV Antibodies/blood , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Humans , Injections, Intramuscular , Interferon-gamma/biosynthesis , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/immunology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vesicular stomatitis Indiana virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...