Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 21(7): 1432-44, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23670573

RESUMO

Marburg and Ebola hemorrhagic fevers have been described as the most virulent viral diseases known to man due to associative lethality rates of up to 90%. Death can occur within days to weeks of exposure and there is currently no licensed vaccine or therapeutic. Recent evidence suggests an important role for antiviral T cells in conferring protection, but little detailed analysis of this response as driven by a protective vaccine has been reported. We developed a synthetic polyvalent-filovirus DNA vaccine against Marburg marburgvirus (MARV), Zaire ebolavirus (ZEBOV), and Sudan ebolavirus (SUDV). Preclinical efficacy studies were performed in guinea pigs and mice using rodent-adapted viruses, whereas murine T-cell responses were extensively analyzed using a novel modified assay described herein. Vaccination was highly potent, elicited robust neutralizing antibodies, and completely protected against MARV and ZEBOV challenge. Comprehensive T-cell analysis revealed cytotoxic T lymphocytes (CTLs) of great magnitude, epitopic breadth, and Th1-type marker expression. This model provides an important preclinical tool for studying protective immune correlates that could be applied to existing platforms. Data herein support further evaluation of this enhanced gene-based approach in nonhuman primate studies for in depth analyses of T-cell epitopes in understanding protective efficacy.


Assuntos
Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/imunologia , Animais , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Immunoblotting , Marburgvirus/imunologia , Marburgvirus/patogenicidade , Camundongos Endogâmicos C57BL , Vacinas de DNA/uso terapêutico , Vacinas Virais/imunologia , Vacinas Virais/uso terapêutico
2.
Mol Ther ; 18(9): 1714-23, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20571540

RESUMO

Type III/lambda interferons (IFNs) were discovered less than a decade ago and are still in the process of being characterized. Although previous studies have focused on the function of IFN-lambda 3 (also known as interleukin (IL)-28B) in a small animal model, it is unknown whether these functions would translate to a larger, more relevant model. Thus in the present study, we have used DNA vaccination as a method of studying the influence of IFN-lambda 3 on adaptive immune responses in rhesus macaques. Results of our study show for the first time that IFN-lambda 3 has significant influence on antigen-specific CD8(+) T-cell function, especially in regards to cytotoxicity. Peripheral CD8(+) T cells from animals that were administered IFN-lambda 3 showed substantially increased cytotoxic responses as gauged by CD107a and granzyme B coexpression as well as perforin release. Moreover, CD8(+) T cells isolated from the mesenteric lymph nodes (MLN) of animals receiving IFN-lambda 3 loaded significant amounts of granzyme B upon extended antigenic stimulation and induced significantly more granzyme B-mediated cell death of peptide pulsed targets. These data suggest that IFN-lambda 3 is a potent effector of the immune system with special emphasis on CD8(+) T-cell killing functions which warrants further study as a possible immunoadjuvant.


Assuntos
Granzimas/metabolismo , Interferons/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , ELISPOT , Citometria de Fluxo , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Macaca
3.
Hum Vaccin ; 7 Suppl: 120-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21266849

RESUMO

Prostate cancer (PCa) remains a significant public health problem. Current treatment modalities for PCa can be useful, but may be accompanied by deleterious side effects and often do not confer long-term control. Accordingly, additional modalities, such as immunotherapy, may represent an important approach for PCa treatment. The identification of tissue-specific antigens engenders PCa an attractive target for immunotherapeutic approaches. Delivery of DNA vaccines with electroporation has shown promising results for prophylactic and therapeutic targets in a variety of species including humans. Application of this technology for PCa immunotherapy strategies has been limited to single antigen and epitope targets. We sought to test the hypothesis that a broader collection of antigens would improve the breadth and effectiveness of a PCa immune therapy approach. We therefore developed highly optimized DNA vaccines encoding prostate-specific antigen (PSA) and prostate-specific membrane antigen (PSMA) as a dual antigen approach to immune therapy of PCa. PSA-and PSMA-specific cellular immunogenicity was evaluated in a mouse model for co-delivery and single antigen vaccination. Mice received 2 immunizations spaced 2 weeks apart and immunogenicity was evaluated 1 week after the second vaccination. Both the PSA and PSMA vaccines induced robust antigen-specific IFNγ responses by ELISpot. Further characterization of cellular immunogenicity by flow cytometry indicated strong antigen-specific TNFα production by CD4+ T cells and IFNγ and IL-2 secretion by both CD4+ and CD8+ T cells. There was also a strong humoral response as determined by PSA-specific seroconversion. These data support further study of this novel approach to immune therapy of PCa.


Assuntos
Eletroporação/métodos , Imunoterapia/métodos , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/imunologia , Neoplasias da Próstata/terapia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , ELISPOT , Feminino , Imunização Secundária/métodos , Interferon gama/metabolismo , Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Doenças dos Roedores/terapia , Fator de Necrose Tumoral alfa/metabolismo , Vacinação/métodos , Vacinas de DNA/genética
4.
Cytometry A ; 77(3): 275-84, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20104580

RESUMO

The capacity for robust proliferation upon re-infection is a hallmark of adaptive immunity and the basis of vaccination. A widely used animal model for the study of human disease is the rhesus macaque (RM), where capacity for proliferation can be assessed ex vivo using carboxyfluorescein succinimidyl ester (CFSE)-based dilution assays. However, we show over the course of the standard ex vivo proliferation assay that CFSE-labeling at commonly used dye concentrations induces significant cell death, but that this phenomenon is dose-dependent. Here, we describe an alternative semiquantitative method for estimating T cell proliferative responses that avoids the putative biases associated with chemical modification. RM peripheral blood mononuclear cells were stimulated ex vivo with cognate peptides for 5 days, immunostained for intracellular Ki-67, and then analyzed by flow cytometry. We describe a gating strategy using Ki-67 and side light scatter, also a marker of blastogenesis, which correlates strongly with data from CFSE dilution. We show that this method is a valid tool for measuring RM antigen-specific cellular proliferation ex vivo and can be used as an alternative to CFSE dilution assays.


Assuntos
Citometria de Fluxo/métodos , Antígeno Ki-67/biossíntese , Linfócitos T/citologia , Animais , Antígenos/química , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Corantes/farmacologia , Relação Dose-Resposta a Droga , Fluoresceínas/química , Deleção de Genes , Leucócitos Mononucleares/citologia , Macaca mulatta , Succinimidas/química , Linfócitos T/microbiologia
5.
Expert Rev Vaccines ; 12(5): 537-54, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23659301

RESUMO

The human body has developed an elaborate defense system against microbial pathogens and foreign antigens. However, particular microbes have evolved sophisticated mechanisms to evade immune surveillance, allowing persistence within the human host. In an effort to combat such infections, intensive research has focused on the development of effective prophylactic and therapeutic countermeasures to suppress or clear persistent viral infections. To date, popular therapeutic strategies have included the use of live-attenuated microbes, viral vectors and dendritic-cell vaccines aiming to help suppress or clear infection. In recent years, improved DNA vaccines have now re-emerged as a promising candidate for therapeutic intervention due to the development of advanced optimization and delivery technologies. For instance, genetic optimization of synthetic plasmid constructs and their encoded antigens, in vivo electroporation-mediated vaccine delivery, as well as codelivery with molecular adjuvants have collectively enhanced both transgene expression and the elicitation of vaccine-induced immunity. In addition, the development of potent heterologous prime-boost regimens has also provided significant contributions to DNA vaccine immunogenicity. Herein, the authors will focus on these recent improvements to this synthetic platform in relation to their application in combating persistent virus infection.


Assuntos
DNA/imunologia , Vacinas Virais/imunologia , Viroses/imunologia , Viroses/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Antígenos Virais/genética , Antígenos Virais/imunologia , Doença Crônica , DNA/administração & dosagem , DNA/genética , Descoberta de Drogas/tendências , Eletroporação , Humanos , Plasmídeos , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
6.
Hum Vaccin Immunother ; 8(11): 1668-81, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23151448

RESUMO

There is no licensed vaccine or cure for human cytomegalovirus (CMV), a ubiquitous ß-herpesvirus infecting 60-95% of adults worldwide. Infection can cause congenital abnormalities, result in severe disease in immunocompromised patients, and is a major impediment during successful organ transplantation. In addition, it has been associated with numerous inflammatory diseases and cancers, as well as being implicated in the development of essential hypertension, a major risk factor for heart disease. To date, limited data regarding the identification of immunogenic viral targets has frustrated CMV vaccine development. Based upon promising clinical data suggesting an important role for T cells in protecting against disease in the transplantation setting, we designed a novel panel of highly-optimized synthetic vaccines encoding major CMV proteins and evaluated their immune potential in murine studies. Vaccination induced robust CD8+ and CD4+ T cells of great epitopic breadth as extensively analyzed using a novel modified T cell assay described herein. Together with improved levels of CMV-specific T cells as driven by a vaccine, further immune evaluation of each target is warranted. The present model provides an important tool for guiding future immunization strategies against CMV.


Assuntos
Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/imunologia , Linfócitos T/imunologia , Vacinas Sintéticas/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Eletroporação , Feminino , Citometria de Fluxo , Terapia Genética , Camundongos , Camundongos Endogâmicos C57BL
7.
Vaccine ; 29(39): 6755-62, 2011 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-21238574

RESUMO

Protection against infection is the hallmark of immunity and the basis of effective vaccination. For a variety of reasons there is a great demand to develop new, safer and more effective vaccine platforms. In this regard, while 'first-generation' DNA vaccines were poorly immunogenic, new genetic 'optimization' strategies and the application of in vivo electroporation (EP) have dramatically boosted their potency. We developed a highly optimized plasmid DNA vaccine that expresses the lymphocytic choriomeningitis virus (LCMV) nucleocapsid protein (NP) and evaluated it using the LCMV challenge model, a gold standard for studying infection and immunity. When administered intramuscularly with EP, robust NP-specific cellular and humoral immune responses were elicited, the magnitudes of which approached those following acute LCMV infection. Furthermore, these responses were capable of providing 100% protection against a high-dose, normally lethal virus challenge. This is the first non-infectious vaccine conferring complete protective immunity up to 8 weeks after vaccination and demonstrates the potential of 'next-generation' DNA vaccines.


Assuntos
Imunidade Celular , Imunidade Humoral , Coriomeningite Linfocítica/prevenção & controle , Proteínas do Nucleocapsídeo/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Formação de Anticorpos , ELISPOT , Feminino , Vetores Genéticos , Células HEK293 , Humanos , Dose Letal Mediana , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , Plasmídeos/metabolismo , Transfecção , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas Virais/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa