Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 184(9): 5360-7, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20308630

RESUMEN

Multiple TLR agonists have been shown to have antitumor effects in animal models. However, the therapeutic efficacy of TLR agonist monotherapy in cancer treatment has been limited, and the mechanisms of failure remain unknown. We demonstrate that topical treatment with a TLR-7 agonist, imiquimod, can elicit significant regression of spontaneous breast cancers in neu transgenic mice, a model of human HER-2/neu(+) breast cancer. However, tumor growth progressed once imiquimod therapy was ended. Gene expression analysis using tumor-derived RNA demonstrated that imiquimod induced high levels of IL-10 in addition to TNF-alpha and IFN-gamma. Elevated levels of circulating IL-10 were also detected in sera from imiquimod-treated mice. Elevated serum IL-10 appeared to be derived from IL-10 and dual cytokine secreting (IFN-gamma(+) and IL-10(+)) CD4(+) T cells rather than CD4(+)CD25(+)Foxp3(+) T regulatory cells, which were also induced by imiquimod treatment. Blockade of IL-10, but not TGF-beta, enhanced the antitumor effect of imiquimod by significantly prolonging survival in treated mice. These data suggest that the excessive inflammation induced by TLR agonists may result in a self-regulatory immunosuppression via IL-10 induction and that blocking IL-10 could enhance the therapeutic efficacy of these agents.


Asunto(s)
Mediadores de Inflamación/fisiología , Interleucina-10/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Glicoproteínas de Membrana/agonistas , Invasividad Neoplásica/patología , Receptor Toll-Like 7/agonistas , Enfermedad Aguda , Administración Tópica , Aminoquinolinas/metabolismo , Aminoquinolinas/uso terapéutico , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Inhibidores de Crecimiento/agonistas , Inhibidores de Crecimiento/metabolismo , Inhibidores de Crecimiento/uso terapéutico , Imiquimod , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/uso terapéutico , Interleucina-10/sangre , Ligandos , Neoplasias Mamarias Experimentales/inmunología , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapéutico , Ratones , Ratones Transgénicos , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/prevención & control , Distribución Aleatoria , Receptor ErbB-2/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 7/uso terapéutico , Insuficiencia del Tratamiento , Regulación hacia Arriba/inmunología
2.
Mol Ther ; 19(3): 594-601, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21157438

RESUMEN

DNA vaccination is an attractive approach to induce antigen-specific cytotoxic CD8(+) T lymphocytes (CTLs), which can mediate protective antitumor immunity. The potency of DNA vaccines encoding weakly immunogenic tumor-associated antigens (TAAs) can be enhanced by codelivering gene-encoded adjuvants. Pattern recognition receptors (PRRs) that sense intracellular DNA could potentially be used to harness intrinsic immune-stimulating properties of plasmid DNA vaccines. Consequently, the cytosolic DNA sensor, DNA-dependent activator of interferon (IFN) regulatory factors (DAI), was used as a genetic adjuvant. In vivo electroporation (EP) of mice with a DAI-encoding plasmid (pDAI) promoted transcription of genes encoding type I IFNs, proinflammatory cytokines, and costimulatory molecules. Coimmunization with pDAI and antigen-encoding plasmids enhanced in vivo antigen-specific proliferation, and induction of effector and memory CTLs. Moreover, codelivery of pDAI effectively promoted CTL and CD4(+) Th1 responses to the TAA survivin. The DAI-enhanced CTL induction required nuclear factor κB (NF-κB) activation and type I IFN signaling, but did not involve the IFN regulatory factor 3 (IRF3). Codelivery of pDAI also increased CTL responses to the melanoma-associated antigen tyrosinase-related protein-2 (TRP2), enhanced tumor rejection and conferred long-term protection against B16 melanoma challenge. This study constitutes "proof-of-principle" validating the use of intracellular PRRs as genetic adjuvants to enhance DNA vaccine potency.


Asunto(s)
Adyuvantes Inmunológicos/genética , Glicoproteínas , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN , Inmunidad Adaptativa/genética , Animales , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Glicoproteínas/inmunología , Inmunidad Innata/genética , Proteínas Inhibidoras de la Apoptosis/inmunología , Oxidorreductasas Intramoleculares/inmunología , Activación de Linfocitos/genética , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plásmidos/genética , Plásmidos/inmunología , Proteínas de Unión al ARN , Proteínas Represoras/inmunología , Análisis de Supervivencia , Survivin , Vacunas de ADN/inmunología
3.
Cancer Immunol Immunother ; 59(1): 81-92, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19526360

RESUMEN

Survivin is an intracellular tumor-associated antigen that is broadly expressed in a large variety of tumors and also in tumor associated endothelial cells but mostly absent in differentiated tissues. Naked DNA vaccines targeting survivin have been shown to induce T cell as well as humoral immune responses in mice. However, the lack of epitope-specific CD8+ T cell detection and modest tumor protection observed highlight the need for further improvements to develop effective survivin DNA vaccination approaches. Here, the efficacy of a human survivin DNA vaccine delivered by intradermal electroporation (EP) was tested. The CD8+ T cell epitope surv(20-28) restricted to H-2 Db was identified based on in-silico epitope prediction algorithms and binding to MHC class I molecules. Intradermal DNA EP of mice with a human survivin encoding plasmid generated CD8+ cytotoxic T lymphocyte (CTL) responses cross-reactive with the mouse epitope surv(20-28), as determined by intracellular IFN-gamma staining, suggesting that self-tolerance has been broken. Survivin-specific CTLs displayed an activated effector phenotype as determined by CD44 and CD107 up-regulation. Vaccinated mice displayed specific cytotoxic activity against B16 and peptide-pulsed RMA-S cells in vitro as well as against surv(20-28) peptide-pulsed target cells in vivo. Importantly, intradermal EP with a survivin DNA vaccine suppressed angiogenesis in vivo and elicited protection against highly aggressive syngeneic B16 melanoma tumor challenge. We conclude that intradermal EP is an attractive method for delivering a survivin DNA vaccine that should be explored also in clinical studies.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Melanoma Experimental/inmunología , Proteínas Asociadas a Microtúbulos/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/administración & dosificación , Animales , Vacunas contra el Cáncer/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Electroporación , Humanos , Proteínas Inhibidoras de la Apoptosis , Inyecciones Intradérmicas , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inmunología , Neovascularización Patológica/terapia , Survivin , Vacunas de ADN/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA