Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Transl Med ; 4: 1, 2006 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-16390546

RESUMO

PURPOSE: Based on previous studies that demonstrated the safety profile and preliminary clinical activity of prostate specific antigen (PSA) targeted therapeutic vaccines, as well as recent laboratory data supporting the value of the addition of co-stimulatory molecules B7-1, ICAM-1, and LFA-3 (designated TRICOM) to these vaccines, we conducted a Phase I study to evaluate the safety and immunogenicity of a novel vaccinia and fowlpox vaccine incorporating the PSA gene sequence and TRICOM. METHODS: In this study, ten patients with androgen independent prostate cancer with or without metastatic disease were enrolled. Patients were treated with 2 x l0(8) pfu of a recombinant vaccinia virus vaccine (PROSTVAC-V) followed by 1 x 10(9) pfu of the booster recombinant fowlpox virus (PROSTVAC-F) both with gene sequences for PSA and TRICOM. The mean age of patients enrolled in the study was 70 (range 63 to 79). The mean PSA at baseline was 434 (range 9-1424). RESULTS: There were no deaths, and no Grade 3 or 4 adverse events. The most commonly reported adverse events, regardless of causality, were injection site reactions and fatigue. One serious adverse event (SAE) occurred that was unrelated to vaccine; this patient developed progressive disease with a new sphenoid metastasis. PSA was measured at week 4 and week 8. Four patients had stable disease (with less than 25% increase in PSA) through the week 8 study period. Anti-PSA antibodies were not induced with therapy: however, anti-vaccinia titers increased in all patients. CONCLUSION: This study demonstrated that vaccination with PROSTVAC-V and PROSTVAC-F combined with TRICOM is well-tolerated and generated an immune response to vaccinia. Therefore, PROSTVAC-VF/TRICOM represents a feasible therapeutic approach for further phase II and III study in patients with prostate cancer.

2.
J Med Primatol ; 31(4-5): 179-85, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12390540

RESUMO

We compared the immunogenicity of plasmid vaccines containing multiple human immunodeficiency virus (HIV) antigens and found that covaccination with plasmids expressing HIV-1 14 kDa vpr gene product profoundly reduces antigen-specific CD8-mediated cytotoxic T-cell activity (CTL). Interestingly, Th1 type responses against codelivered antigens (pGag-Pol, pNef, etc.) encoded by the plasmid vaccines were suppressed. This suggested that vpr might compromise CD8 T-cell immunity in vivo during infection. A pilot primate vaccine study was designed to test the hypothesis to compare the following groups: unvaccinated controls, animals vaccinated without simean immunodeficiency virus (SIV)-Nef antigen plasmid, and animals covaccinated with the identical plasmid antigen and a plasmid construct encoding SIV Vpr/Vpx. Animals were subsequently challenged intrarectally with pathogenic SIVmac251 after the final vaccination of a multiple immunization protocol. Control animals were all infected and exhibited high viral loads and rapid CD4+ T-cell loss. In contrast, the Nef plasmid-vaccinated animals were also infected but exhibited preservation of CD4+ T-cells and a multilog reduction in viral load compared with controls. Animals covaccinated multiple times with the Nef vaccine and pVpr/Vpx plasmid suffered rapid and profound loss of CD4+ T-cells. These results have important implications for the design of multicomponent and particle vaccines for HIV-1 as well as for our understanding of HIV/SIV pathogenesis in vivo.


Assuntos
Linfócitos T CD4-Positivos/citologia , Produtos do Gene nef/imunologia , Produtos do Gene vpr/imunologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Vacinas contra a SAIDS/genética , Vacinas contra a SAIDS/imunologia , Vírus da Imunodeficiência Símia/imunologia , Carga Viral , Animais , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Produtos do Gene nef/genética , Produtos do Gene vpr/genética , Modelos Logísticos , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , RNA Viral/sangue , RNA Viral/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/fisiologia , Fatores de Tempo , Vacinas de DNA/genética , Vacinas de DNA/imunologia
3.
Int J Parasitol ; 32(5): 543-50, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11943227

RESUMO

It is widely believed that a Th1 type CD4 response is critical for enhancement of CD8 immunity and for controlling HIV-1 infection. Th2 type responses, such as what might be seen in a chronic parasitic infection, would sacrifice cellular immunity and thus benefit the virus at the expense of the host. However, there has been little direct examination of the hypothesis in a primate model system. Accordingly, the simian immunodeficiency virus (SIV) infected rhesus macaque model was used to investigate the impact of immunisation with SIV expressing DNA constructs and co-injection with IL-4 on the SIV specific immunological responses, lymphocyte cell counts, as well as the impact on viral load. IL-4 is a Th2 type cytokine, which enhances antibody production and inhibits a CD4 Th1 phenotype. Rhesus macaques were infected with 10 AID50 of SIVmac239 and treated with 9-[2-(phosphonomethoxy)propyl]adenine (PMPA) 9 weeks post-infection. During PMPA treatment, animals were immunised with plasmids that expressed the SIV proteins, env, rev, gag and pol. In addition, they were immunised with a construct that encoded the gene for IL-4. IL-4 co-immunisation increased the neutralizing antibody titres in this group. Importantly, the viral loads in animals vaccinated with IL-4 expressing plasmid increased during the immunisation regimens despite the higher neutralizing antibody titres. In addition, neutralizing antibodies did not correlate with viral set point prior to PMPA treatment, however, there was a correlation between viral loads and antibody titres following the treatment with PMPA. Antibody titres decreased following the suppression of viral load. Importantly, vaccination in the absence of IL-4 protected CD4 levels without increasing viral load. The data support the hypothesis that inappropriate immune bias toward a Th2 pathway would ultimately enhance disease progression.


Assuntos
Interleucina-4/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral , Animais , Anticorpos Antivirais/sangue , Contagem de Linfócito CD4 , Humanos , Imunização , Interleucina-4/administração & dosagem , Interleucina-4/genética , Ativação Linfocitária , Macaca mulatta , Plasmídeos/genética , Vacinas contra a SAIDS/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Carga Viral
4.
Oncogene ; 20(33): 4497-506, 2001 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-11494145

RESUMO

Prostate specific antigen (PSA) is a widely used marker for prostate cancer, which is secreted by normal prostate cells at low levels, but is produced more substantially by cancer cells. We have previously reported on the use of a DNA vaccine construct that encodes for human PSA gene to elicit host immune responses against cells producing PSA. DNA immunization strategy delivers DNA constructs encoding for a specific immunogen into the host, who becomes the in vivo protein source for the production of antigen. This antigen then is the focus of the resulting immune response. In this study, we examine the induction of immune responses and safety profiles in rhesus macaques immunized with DNA-based PSA vaccine. We observed induction of PSA-specific humoral response as well as positive PSA-specific lymphoproliferative (LPA) response in the vaccinated macaques. We also observed that the stimulated T cells from the PSA-immunized rhesus macaques produced higher levels of Th1 type cytokine IFN-gamma than the control vector immunized animals. On the other hand, DNA immunization did not result in any adverse effects in the immunized macaques, as indicated by complete blood counts, leukocyte differentials and hepatic and renal chemistries. The macaques appeared healthy, without any physical signs of toxicity throughout the observation period. In addition, we did not observe any adverse effect on the vaccination site. The apparent safety and immunogenecity of DNA immunization in this study suggest that further evaluation of this vaccination strategy is warranted.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Antígeno Prostático Específico/imunologia , Vacinas de DNA/imunologia , Animais , Contagem de Células Sanguíneas , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/toxicidade , Relação Dose-Resposta Imunológica , Humanos , Interferon gama/metabolismo , Testes de Função Renal , Testes de Função Hepática , Ativação Linfocitária , Macaca mulatta , Masculino , Próstata/efeitos dos fármacos , Segurança , Células Th1/imunologia , Células Th1/metabolismo , Vacinação/efeitos adversos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/toxicidade , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/toxicidade
5.
Virology ; 285(2): 204-17, 2001 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-11437655

RESUMO

Multicomponent DNA vaccines were used to elicit immune responses, which can impact viral challenge in three separate rhesus macaque models. Eight rhesus macaques were immunized with DNA vaccines for HIV env/rev and SIV gag/pol and were challenged intravenously with 10 animal infective doses (AID(50)) of cell-free SHIV IIIB. Three of eight immunized rhesus macaques were protected, exhibiting no detectable virus. Animals protected from nonpathogenic SHIVIIIB challenge were rested for extended periods of time and were rechallenged first with pathogenic SIV(mac239) and subsequently with pathogenic SHIV89.6P viruses. Following the pathogenic challenges, all three vaccinated animals were negative for viral coculture and antigenemia and were negative by PCR. In contrast, the control animals exhibited antigenemia by 2 weeks postchallenge and exhibited greater than 10 logs of virus/10(6) cells in limiting dilution coculture. The control animals exhibited CD4 cell loss and developed SIV-related wasting with high viral burden and subsequently failed to thrive. Vaccinated animals remained virus-negative and were protected from the viral load, CD4 loss, disease, and death. We observed strong Th1-type cellular immune responses in the protected macaques throughout the study, suggesting their important roles in protection. These studies support the finding that multicomponent DNA vaccines can directly impact viral replication and disease in a highly pathogenic challenge system, thus potentially broadening our strategies against HIV.


Assuntos
Vacinas contra a AIDS/imunologia , HIV-1/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Vacinas de DNA/imunologia , Animais , HIV-1/genética , Humanos , Macaca mulatta
6.
Clin Cancer Res ; 7(3 Suppl): 882s-889s, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11300487

RESUMO

DNA immunization is an important vaccination technique that is being explored as an immunotherapeutic strategy against a variety of infectious diseases as well as cancer. We have been investigating the utility of DNA-based vaccine strategy against prostate cancer. We have developed a DNA vaccine construct that encodes for the human prostate specific antigen (PSA) gene. PSA expression is limited to prostate cells, and the level of PSA expression is substantially increased in prostate cancer cells. This tissue specificity makes PSA a potential target for the development of immunotherapies against prostate cancer. A DNA-based PSA vaccine was used to elicit PSA-specific host immune responses in rodent and nonhuman primate models. In an effort to enhance the clinical utility of the DNA-based PSA vaccine, we also examined the use of cytokine gene adjuvants to modulate vaccine-induced immune responses in these animal models. We observed that pCPSA vaccine-induced humoral and cellular immune responses can be modulated through the coimmunization with cytokine genes in mice, and these enhancement effects on the PSA-specific cellular responses were extended in macaques. More specifically, coimmunization with interleukin (IL)-2 cDNA construct resulted in a significant enhancement of PSA-specific antibody responses in both mice and macaque models. In contrast, coinjection of IL-12 resulted in reduction of antibody responses in both models. In mice, the groups coimmunized with IL-2, IL-12, or IL-18 showed a dramatic increase in T helper cell proliferation over the results with pCPSA alone. These results support that further evaluation of this vaccination strategy to treat prostate cancer is warranted.


Assuntos
Vacinas Anticâncer , Citocinas/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Vacinas de DNA , Animais , Divisão Celular , DNA Complementar/metabolismo , Ensaio de Imunoadsorção Enzimática , Interleucina-12/uso terapêutico , Interleucina-18/uso terapêutico , Interleucina-2/uso terapêutico , Linfócitos/metabolismo , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Antígeno Prostático Específico/genética , Linfócitos T/metabolismo , Fatores de Tempo
7.
J Urol ; 165(4): 1300-4, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11257705

RESUMO

PURPOSE: We determined whether the cytotoxicity of doxorubicin hydrochloride would be enhanced by adding hydrogen peroxide as a source of oxygen free radicals. MATERIALS AND METHODS: Mouse bladder tumor cells (MBT-2) were grown in RPMI 1640 medium and treated with various concentrations of doxorubicin hydrochloride for 2 hours. Protein content was assayed as a measure of cell growth. A similar set of experiments was done with cells exposed to hydrogen peroxide only and combined doxorubicin and hydrogen peroxide. Protein content was again assayed as a measure of cell growth. Cells were also assayed for glutathione peroxidase and malonyl dialdehyde, a product of lipid peroxidation, to determine the mechanism of cell damage. Furthermore, MBT-2 cells were incubated with 100 M. alpha-tocopherol, a free radical scavenger, before exposure to hydrogen peroxide to determine whether the effects of hydrogen peroxide could be reversed. RESULTS: We observed a dose dependent inhibition of MBT-2 cell growth after exposure to doxorubicin hydrochloride. Exposure to doxorubicin and hydrogen peroxide resulted in greater cell growth inhibition than exposure to either agent alone. The effects of hydrogen peroxide on cell proliferation were reversed by pre-incubation with alpha-tocopherol. CONCLUSIONS: As a source of oxygen free radicals, hydrogen peroxide enhances the antiproliferative effect of doxorubicin hydrochloride on a mouse bladder tumor cell line. Thus, hydrogen peroxide may be a relatively inexpensive, nontoxic method of augmenting the cytotoxicity of doxorubicin hydrochloride. Further studies are warranted to determine whether these observations may have clinical application.


Assuntos
Antineoplásicos/uso terapêutico , Doxorrubicina/uso terapêutico , Peróxido de Hidrogênio/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Sequestradores de Radicais Livres , Peroxidação de Lipídeos , Camundongos , Camundongos Endogâmicos C3H , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/fisiopatologia
8.
Vaccine ; 19(17-19): 2496-505, 2001 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-11257383

RESUMO

Extensive experiments have shown DNA vaccines' ability to elicit immune responses in vivo in a safe and well-tolerated manner in several model systems, including rodents and non-human primates. As the DNA-based vaccine and immunotherapy approaches are being explored in humans, significant efforts have also been focused on further improving the immune potency of this technology. One strategy to enhance immune responses for DNA vaccines is the use of molecular or genetic adjuvants. These molecular adjuvant constructs (which encodes for immunologically important molecules such as cytokines) can be co-administered along with DNA vaccine constructs. Once delivered, these adjuvants have shown to modulate the magnitude and direction (humoral or cellular) of the vaccine-induced immune responses in rodent models. To date, however, there has been very little data reported from studies in primates. In this study, we examined the effects of cytokine gene adjuvants to enhance the level of cell-mediated immune responses in rhesus macaques. We co-immunized rhesus macaques with expression plasmids encoding for IL-2, IFN-gamma or IL-4 cytokines along with the DNA vaccine constructs encoding for HIV env/rev (pCEnv) and SIV gag/pol (pCSGag/pol) proteins. We observed that coadministration of IL-2 and IFN-gamma cDNA resulted in enhancement of antigen-specific T cell-mediated immune responses.


Assuntos
Adjuvantes Imunológicos/genética , Citocinas/genética , Imunidade Celular , Vacinas de DNA/farmacologia , Vacinas contra a AIDS/genética , Vacinas contra a AIDS/farmacologia , Animais , Antígenos Virais/genética , Expressão Gênica , Humanos , Técnicas In Vitro , Interferon gama/genética , Interleucina-2/genética , Interleucina-4/genética , Ativação Linfocitária , Macaca mulatta , Vacinas contra a SAIDS/genética , Vacinas contra a SAIDS/farmacologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/genética
9.
Vaccine ; 19(13-14): 1738-46, 2001 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-11166899

RESUMO

HIV-1 Tat, a secreted virally encoded toxin, enhances chronic viral replication and induces immune suppression, activities blocked in vitro and in vivo by anti-Tat antibodies. We mapped HIV-1 Tat B cell epitopes, determined sequence variation within them in 350 Tat sequences in GenBank, and determined antigenic cross-reactions between significant amino acid polymorphs. Two of the four B cell epitope sequences identified had limited or no antigenic polymorphism within geographically diverse strains. For epitope 1 in primates, (V,I)4DP(R,K,S,N)7L(E,D)9PW(N,K)12, the most frequent antigenic polymorphs were VDPRLEPWK in B clades (75%) and VDPNLEPWN in non-B clades (64%), with five additional sequences occurring at lower incidence. Epitope 2 in primates, K41(G,A)42LGISYGRK50, had no antigenic polymorphism. These two epitopes have potential utility for the generation of universal vaccine immunogens and therapeutic antibodies.


Assuntos
Epitopos de Linfócito B/genética , Epitopos de Linfócito B/imunologia , Produtos do Gene tat/imunologia , HIV-1/genética , HIV-1/imunologia , Polimorfismo Genético/genética , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Linfócitos B/imunologia , Sequência Consenso/genética , Sequência Conservada/genética , Reações Cruzadas/imunologia , Análise Mutacional de DNA , Bases de Dados como Assunto , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Produtos do Gene tat/química , Produtos do Gene tat/genética , Variação Genética/genética , Anticorpos Anti-HIV/imunologia , HIV-1/química , HIV-1/classificação , Haplorrinos/imunologia , Soros Imunes/imunologia , Dados de Sequência Molecular , Mutação/genética , Coelhos , Produtos do Gene tat do Vírus da Imunodeficiência Humana
10.
J Virol ; 75(1): 73-82, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11119575

RESUMO

Cytotoxic T-lymphocyte (CTL) responses have been implicated as playing an important role in control of human immunodeficiency virus (HIV) infection. However, it is technically difficult to demonstrate CTL responses consistently in nonhuman primate and human subjects using traditional cytotoxicity assay methods. In this study, we systematically evaluated culture conditions that may affect the proliferation and expansion of CTL effector cells and presented a sensitive method for detection of cytotoxicity responses with bulk CTL cultures. We confirmed the sensitivity and specificity of this method by demonstration of vigorous CTL responses in a simian-HIV (SHIV)-infected rhesus macaque. The expansion of epitope-specific CTL effector cells was also measured quantitatively by CTL epitope-major histocompatibility complex tetramer complex staining. In addition, two new T-cell determinants in the SIV gag region are identified. Last, we showed the utility of this method for studying CTL responses in chimpanzee and human subjects.


Assuntos
Síndrome da Imunodeficiência Adquirida/imunologia , HIV-1 , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Humanos , Interleucina-7/farmacologia , Macaca mulatta
11.
J Virol ; 74(22): 10514-22, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11044096

RESUMO

We report a pilot evaluation of a DNA vaccine producing genetically inactivated simian immunodeficiency virus (SIV) particles in primates, with a focus on eliciting mucosal immunity. Our results demonstrate that DNA vaccines can be used to stimulate strong virus-specific mucosal immune responses in primates. The levels of immunoglobulin A (IgA) detected in rectal secretions of macaques that received the DNA vaccine intradermally and at the rectal mucosa were the most striking of all measured immune responses and were higher than usually achieved through natural infection. However, cytotoxic T lymphocyte responses were generally low and sporadically present in different animals. Upon rectal challenge with cloned SIVmac239, resistance to infection was observed, but some animals with high SIV-specific IgA levels in rectal secretions became infected. Our results suggest that high levels of IgA alone are not sufficient to prevent the establishment of chronic infection, although mucosal IgA responses may have a role in reducing the infectivity of the initial viral inoculum.


Assuntos
Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas Atenuadas/imunologia , Vacinas de DNA/imunologia , Animais , Antígenos Virais/imunologia , DNA Viral/genética , Imunidade nas Mucosas , Imunoglobulina A/análise , Macaca mulatta , Provírus/genética , Vacinas contra a SAIDS/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Linfócitos T Citotóxicos/imunologia , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas de DNA/administração & dosagem , Vírion/genética , Vírion/fisiologia
12.
Antimicrob Agents Chemother ; 44(11): 3199-202, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11036053

RESUMO

Human immunodeficiency virus type 1 (HIV-1) infection continues to spread in developing countries, mostly through heterosexual transmission. The development of a safe and cost-effective topical microbicide, effective against a range of STDs including HIV-1, would greatly impact the ongoing epidemic. When formulated in a vehicle, a micronized form of cellulose acetate phthalate (CAP), which is an inactive pharmaceutical excipient, has been shown to inactivate HIV-1, herpes simplex virus types 1 and 2, cytomegalovirus, Neisseria gonorrhoeae, Trichomonas vaginalis, Haemophilus ducreyi, and Chlamydia trachomatis in vitro. Formulated CAP was also shown to be effective against herpes simplex virus type 2 in vivo. Here we show that a formulation of CAP protected four of six rhesus monkeys from vaginal infection with simian immunodeficiency virus. Thus, CAP may be a candidate for use as a topical microbicide for preventing HIV-1 infection in humans.


Assuntos
Antivirais/uso terapêutico , Celulose/análogos & derivados , Celulose/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Administração Tópica , Animais , Antivirais/administração & dosagem , Celulose/administração & dosagem , Modelos Animais de Doenças , Transmissão de Doença Infecciosa/prevenção & controle , Infecções por HIV/transmissão , Humanos , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/fisiologia , Cremes, Espumas e Géis Vaginais
13.
J Virol ; 74(16): 7485-95, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10906202

RESUMO

The potential contribution of a plasmid DNA construct to vaccine-elicited protective immunity was explored in the simian immunodeficiency virus (SIV)/macaque model of AIDS. Making use of soluble major histocompatibility class I/peptide tetramers and peptide-specific killing assays to monitor CD8(+) T-lymphocyte responses to a dominant SIV Gag epitope in genetically selected rhesus monkeys, a codon-optimized SIV gag DNA vaccine construct was shown to elicit a high-frequency SIV-specific cytotoxic T-lymphocyte (CTL) response. This CTL response was demonstrable in both peripheral blood and lymph node lymphocytes. Following an intravenous challenge with the highly pathogenic viral isolate SIVsm E660, these vaccinated monkeys developed a secondary CTL response that arose with more rapid kinetics and reached a higher frequency than did the postchallenge CTL response in control plasmid-vaccinated monkeys. While peak plasma SIV RNA levels were comparable in the experimentally and control-vaccinated monkeys during the period of primary infection, the gag plasmid DNA-vaccinated monkeys demonstrated better containment of viral replication by 50 days following SIV challenge. These findings indicate that a plasmid DNA vaccine can elicit SIV-specific CTL responses in rhesus monkeys, and this vaccine-elicited immunity can facilitate the generation of secondary CTL responses and control of viral replication following a pathogenic SIV challenge. These observations suggest that plasmid DNA may prove a useful component of a human immunodeficiency virus type 1 vaccine.


Assuntos
Produtos do Gene gag/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/imunologia , Animais , Contagem de Linfócito CD4 , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Humanos , Linfonodos/imunologia , Linfonodos/virologia , Macaca mulatta , RNA Viral/análise , Vacinas contra a SAIDS/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Vacinação , Vacinas de DNA/administração & dosagem , Carga Viral , Replicação Viral
14.
Vaccine ; 18(25): 2789-95, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10812220

RESUMO

HIV-1 Tat protein activates resting cells, rendering them permissive for viral replication. Replication of HIV-1 in vitro is enhanced by intercellular passage of Tat protein and inhibited by anti-Tat antibodies. Tat dependence of HIV-1 replication in vivo during acute, chronic asymptomatic and AIDS stages of infection was assessed by comparisons of plasma viremia in Tat-immunized or control monkeys challenged with SHIV(33) or SHIV(33A). Chronic plasma viremia became undetectable or minimized in Tat-immunized asymptomatic SHIV(33)-infected monkeys (p<0.008) while the high viral loads of acute infection or SHIV(33A)-induced simian AIDS were unaffected by Tat immunization. Active or passive immunotherapies targeting Tat provide potential approaches to controlling chronic HIV-1 viremia and preventing AIDS.


Assuntos
Produtos do Gene tat/imunologia , Infecções por HIV/prevenção & controle , HIV-1/genética , HIV-1/imunologia , Peptídeos/imunologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Viremia/prevenção & controle , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/biossíntese , Contagem de Linfócito CD4 , Quimera , Doença Crônica , Ensaio de Imunoadsorção Enzimática , Produtos do Gene tat/efeitos adversos , Produtos do Gene tat/síntese química , Infecções por HIV/imunologia , Soropositividade para HIV/imunologia , Humanos , Macaca mulatta , Dados de Sequência Molecular , Peptídeos/efeitos adversos , Peptídeos/síntese química , Carga Viral , Produtos do Gene tat do Vírus da Imunodeficiência Humana
15.
J Virol ; 74(7): 3427-9, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10708463

RESUMO

An important limitation of DNA immunization in nonhuman primates is the difficulty in generating high levels of antigen-specific antibody responses; strategies to enhance the level of immune responses to DNA immunization may be important in the further development of this vaccine strategy for humans. We approached this issue by testing the ability of molecular adjuvants to enhance the levels of immune responses generated by multicomponent DNA vaccines in rhesus macaques. Rhesus macaques were coimmunized intramuscularly with expression plasmids bearing genes encoding Th1 (interleukin 2 [IL-2] and gamma interferon)- or Th2 (IL-4)-type cytokines and DNA vaccine constructs encoding human immunodeficiency virus Env and Rev and simian immunodeficiency virus Gag and Pol proteins. We observed that the cytokine gene adjuvants (especially IL-2 and IL-4) significantly enhanced antigen-specific humoral immune responses in the rhesus macaque model. These results support the assumption that antigen-specific responses can be engineered to a higher and presumably more desirable level in rhesus macaques by genetic adjuvants.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Antivirais/biossíntese , Antígenos Virais/imunologia , Citocinas/genética , DNA Complementar/imunologia , Vacinas Virais/imunologia , Animais , Humanos , Macaca mulatta
16.
J Med Primatol ; 28(4-5): 214-23, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10593488

RESUMO

DNA or nucleic acid immunization has been shown to induce both antigen-specific cellular and humoral immune responses in vivo. Moreover, immune responses induced by DNA immunization can be enhanced and modulated by the use of molecular adjuvants. To engineer the immune response in vivo towards more T-helper (Th)1-type cellular responses, we investigated the co-delivery of inteferon (IFN)-gamma, interleukin (IL)-12, and IL-18 genes along with DNA vaccine constructs. We observed that both antigen-specific humoral and cellular immune responses can be modulated through the use of cytokine adjuvants in mice. Most of this work has been performed in rodent models. There has been little confirmation of this technology in primates. We also evaluated the immunomodulatory effects of this approach in rhesus macaques, since non-human primates represent the most relevant animal models for human immunodeficiency virus (HIV) vaccine studies. As in the murine studies, we also observed that each Th1 cytokine adjuvant distinctively regulated the level of immune responses generated. Co-immunization of IFN-gamma and IL-18 in macaques enhanced the level of antigen-specific antibody responses. Similarly, co-delivery of IL-12 and IL-18 also enhanced the level of antigen-specific Th proliferative responses. These results extend this adjuvant strategy in a more relevant primate model and support the potential utility of these molecular adjuvants in DNA vaccine regimens.


Assuntos
Vacinas contra a AIDS , Reações Antígeno-Anticorpo/imunologia , Interleucina-12/genética , Interleucina-18/genética , Vírus da Imunodeficiência Símia/imunologia , Vacinas de DNA , Adjuvantes Imunológicos , Animais , Formação de Anticorpos/imunologia , Modelos Animais de Doenças , Feminino , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Imunidade Celular/imunologia , Imunização/veterinária , Interleucina-12/imunologia , Interleucina-18/imunologia , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Auxiliares-Indutores/imunologia
17.
J Virol ; 73(12): 10199-207, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10559336

RESUMO

Several different strains of simian-human immunodeficiency virus (SHIV) that contain the envelope glycoproteins of either T-cell-line-adapted (TCLA) strains or primary isolates of human immunodeficiency virus type 1 (HIV-1) are now available. One of the advantages of these chimeric viruses is their application to studies of HIV-1-specific neutralizing antibodies in preclinical AIDS vaccine studies in nonhuman primates. In this regard, an important consideration is the spectrum of antigenic properties exhibited by the different envelope glycoproteins used for SHIV construction. The antigenic properties of six SHIV variants were characterized here in neutralization assays with recombinant soluble CD4 (rsCD4), monoclonal antibodies, and serum samples from SHIV-infected macaques and HIV-1-infected individuals. Neutralization of SHIV variants HXBc2, KU2, 89.6, and 89.6P by autologous and heterologous sera from SHIV-infected macaques was restricted to an extent that these viruses may be considered heterologous to one another in their major neutralization determinants. Little or no variation was seen in the neutralization determinants on SHIV variants 89.6P, 89.6PD, and SHIV-KB9. Neutralization of SHIV HXBc2 by sera from HXBc2-infected macaques could be blocked with autologous V3-loop peptide; this was less true in the case of SHIV 89.6 and sera from SHIV 89.6-infected macaques. The poorly immunogenic but highly conserved epitope for monoclonal antibody IgG1b12 was a target for neutralization on SHIV variants HXBc2, KU2, and 89.6 but not on 89.6P and KB9. The 2G12 epitope was a target for neutralization on all five SHIV variants. SHIV variants KU2, 89.6, 89.6P, 89.6PD, and KB9 exhibited antigenic properties characteristic of primary isolates by being relatively insensitive to neutralization in peripheral blood mononuclear cells with serum samples from HIV-1-infected individuals and 12-fold to 38-fold less sensitive to inhibition with recombinant soluble CD4 than TCLA strains of HIV-1. The utility of nonhuman primate models in AIDS vaccine development is strengthened by the availability of SHIV variants that are heterologous in their neutralization determinants and exhibit antigenic properties shared with primary isolates.


Assuntos
Variação Genética , Infecções por HIV/imunologia , HIV-1/imunologia , Vírus da Imunodeficiência Símia/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Anti-HIV/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/sangue , HIV-1/genética , HIV-1/isolamento & purificação , Humanos , Macaca , Dados de Sequência Molecular , Testes de Neutralização , Fragmentos de Peptídeos/imunologia , Recombinação Genética , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/isolamento & purificação , Células Tumorais Cultivadas
18.
J Virol ; 73(10): 8356-63, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10482586

RESUMO

We examined the ability of a live, attenuated deletion mutant of simian immunodeficiency virus (SIV), SIVmac239Delta3, which is missing nef and vpr genes, to protect against challenge by heterologous strains SHIV89.6p and SIVsmE660. SHIV89.6p is a pathogenic, recombinant SIV in which the envelope gene has been replaced by a human immunodeficiency virus type 1 envelope gene; other structural genes of SHIV89.6p are derived from SIVmac239. SIVsmE660 is an uncloned, pathogenic, independent isolate from the same primate lentivirus subgrouping as SIVmac but with natural sequence variation in all structural genes. The challenge with SHIV89.6p was performed by the intravenous route 37 months after the time of vaccination. By the criteria of CD4(+) cell counts and disease, strong protection against the SHIV89.6p challenge was observed in four of four vaccinated monkeys despite the complete mismatch of env sequences. However, SHIV89.6p infection was established in all four previously vaccinated monkeys and three of the four developed fluctuating viral loads between 300 and 10,000 RNA copy equivalents per ml of plasma 30 to 72 weeks postchallenge. When other vaccinated monkeys were challenged with SIVsmE660 at 28 months after the time of vaccination, SIV loads were lower than those observed in unvaccinated controls but the level of protection was less than what was observed against SHIV89.6p in these experiments and considerably less than the level of protection against SIVmac251 observed in previous experiments. These results demonstrate a variable level of vaccine protection by live, attenuated SIVmac239Delta3 against heterologous virus challenge and suggest that even live, attenuated vaccine approaches for AIDS will face significant hurdles in providing protection against the natural variation present in field strains of virus. The results further suggest that factors other than anti-Env immune responses can be principally responsible for the vaccine protection by live, attenuated SIV.


Assuntos
Vacinas contra a SAIDS , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Animais , Deleção de Genes , Genes nef , Genes vpr , Humanos , Macaca mulatta
19.
J Virol ; 73(6): 4952-61, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10233957

RESUMO

Three different deletion mutants of simian immunodeficiency virus (SIV) that vary in their levels of attenuation were tested for the ability to protect against mucosal challenge with pathogenic SIV. Four female rhesus monkeys were vaccinated by intravenous inoculation with SIVmac239Delta3, four with SIVmac239Delta3X, and four with SIVmac239Delta4. These three vaccine strains exhibit increasing levels of attenuation: Delta3 < Delta3X

Assuntos
Vacinas contra a SAIDS/imunologia , Vagina/virologia , Animais , Anticorpos Antivirais/sangue , Contagem de Linfócito CD4 , Feminino , Macaca mulatta , Linfócitos T Citotóxicos/imunologia , Vacinas Atenuadas/imunologia
20.
Nat Med ; 5(5): 526-34, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10229229

RESUMO

Eight different protocols were compared for their ability to raise protection against immunodeficiency virus challenges in rhesus macaques. The most promising containment of challenge infections was achieved by intradermal DNA priming followed by recombinant fowl pox virus booster immunizations. This containment did not require neutralizing antibody and was active for a series of challenges ending with a highly virulent virus with a primary isolate envelope heterologous to the immunizing strain.


Assuntos
Infecções por Lentivirus/imunologia , Infecções por Lentivirus/prevenção & controle , Vacinação , Vacinas de DNA/uso terapêutico , Vacinas Virais/uso terapêutico , Animais , Anticorpos Antivirais/sangue , Vírus da Varíola das Aves Domésticas/genética , Injeções Intradérmicas , Macaca , Testes de Neutralização , RNA Viral/sangue , Linfócitos T Citotóxicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...