Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
J Immunol ; 183(7): 4312-21, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19734207

RESUMEN

Oncolytic virotherapy may mediate antitumor effects via direct oncolysis or immune-mediated tumor regression. Although the ability of oncolytic viruses to generate adaptive antitumor immunity has been characterized, their interactions with the innate immune system are relatively unclear. Using a human in vitro system, this study investigates the innate immunological consequences of reovirus therapy and its potential to activate NK cell-mediated antitumor activity. Dendritic cells (DC) loaded with reovirus-infected human melanoma Mel888 cells (DC-MelReo), but not reovirus-infected tumor cells alone, induced IFN-gamma production within the NK cell population upon coculture with PBMC, in a cell-to-cell contact-dependent manner. DC-MelReo secreted the chemokines CCL2, 3, 4, 5, 7, 8, 11, and CXCL10; these culture supernatants induced NK cell chemotaxis. Coculture of DC-MelReo with purified NK cells induced reciprocal contact-dependent phenotypic DC maturation, while DC-MelReo elicited up-regulation of the activation marker CD69 on NK cells, in a partially contact and partially IL-12 dependent manner. Significantly, DC-MelReo induced NK cell cytotoxicity toward tumor cells by a type I IFN dependent mechanism. These data demonstrate that tumor infection by reovirus can act via DC to induce NK cell recruitment, activation, and cytotoxicity, along with reciprocal DC maturation. These findings suggest that reciprocal DC-NK cell interactions, following reovirus therapy, may play an important role in altering the immune milieu of the tumor microenvironment and mediating tumor regression.


Asunto(s)
Comunicación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Melanoma Experimental/inmunología , Melanoma Experimental/prevención & control , Reoviridae/inmunología , Línea Celular Tumoral , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Células Dendríticas/patología , Humanos , Células Asesinas Naturales/patología , Melanoma Experimental/virología , Viroterapia Oncolítica/métodos , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/prevención & control , Infecciones por Reoviridae/virología
2.
J Immunol ; 181(5): 3108-15, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18713981

RESUMEN

In vivo, dendritic cells (DC) are programmed to orchestrate innate and adaptive immunity in response to pathogen-derived "danger" signals. Under particular circumstances, DC can also be directly cytotoxic against tumor cells, potentially allowing them to release tumor associated Ags from dying cells and then prime antitumor immunity against them. In this study, we describe the innate characteristics of DC (OK-DC) generated in vitro after exposure of immature human myeloid-derived DC to OK432, a penicillin-inactivated and lyophilized preparation of Streptococcus pyrogenes. OK-DC produced proinflammatory cytokines, stimulated autologous T cell proliferation and IFN-gamma secretion, expressed CCR7, and migrated in response to MIP-3beta. Moreover, OK-DC displayed strong, specific cytotoxicity toward tumor cell targets. This cytotoxicity was associated with novel, OK432-induced up-regulation of CD40L on the cell surface of OK-DC, and was absolutely dependent on expression of CD40 on the tumor targets. These data demonstrate that maturation of human DC with OK432, an adjuvant suitable for clinical use, induces direct tumor cell killing by DC, and describes a novel CD40/CD40L-mediated mechanism for specific DC antitumor cytotoxicity.


Asunto(s)
Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Neoplasias/inmunología , Picibanil/farmacología , Ligando de CD40/genética , Muerte Celular , Células Dendríticas/efectos de los fármacos , Humanos , Neoplasias/patología , Regulación hacia Arriba/efectos de los fármacos
3.
Cancer Immunol Res ; 6(10): 1161-1173, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30209061

RESUMEN

Immunotherapy is showing promise for otherwise incurable cancers. Oncolytic viruses (OVs), developed as direct cytotoxic agents, mediate their antitumor effects via activation of the immune system. However, OVs also stimulate antiviral immune responses, including the induction of OV-neutralizing antibodies. Current dogma suggests that the presence of preexisting antiviral neutralizing antibodies in patients, or their development during viral therapy, is a barrier to systemic OV delivery, rendering repeat systemic treatments ineffective. However, we have found that human monocytes loaded with preformed reovirus-antibody complexes, in which the reovirus is fully neutralized, deliver functional replicative reovirus to tumor cells, resulting in tumor cell infection and lysis. This delivery mechanism is mediated, at least in part, by antibody receptors (in particular FcγRIII) that mediate uptake and internalization of the reovirus/antibody complexes by the monocytes. This finding has implications for oncolytic virotherapy and for the design of clinical OV treatment strategies. Cancer Immunol Res; 6(10); 1161-73. ©2018 AACR.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Melanoma Experimental/terapia , Monocitos/inmunología , Viroterapia Oncolítica , Virus Oncolíticos , Reoviridae , Animales , Línea Celular , Chlorocebus aethiops , Femenino , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Ratones Endogámicos C57BL , Receptores de IgG/inmunología
4.
Clin Cancer Res ; 15(13): 4374-4381, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19509134

RESUMEN

PURPOSE: Reovirus is a naturally occurring oncolytic virus in clinical trials. Although tumor infection by reovirus can generate adaptive antitumor immunity, its therapeutic importance versus direct viral oncolysis is undefined. This study addresses the requirement for viral oncolysis and replication, and the relative importance of antitumor immunity and direct oncolysis in therapy. EXPERIMENTAL DESIGN: Nonantigen specific T cells loaded with reovirus were delivered i.v. to C57BL/6 and severe combined immunodeficient mice bearing lymph node and splenic metastases from the murine melanoma, B16ova, with assessment of viral replication, metastatic clearance by tumor colony outgrowth, and immune priming. Human cytotoxic lymphocyte priming assays were done with dendritic cells loaded with Mel888 cells before the addition of reovirus. RESULTS: B16ova was resistant to direct oncolysis in vitro, and failed to support reovirus replication in vitro or in vivo. Nevertheless, reovirus purged lymph node and splenic metastases in C57BL/6 mice and generated antitumor immunity. In contrast, reovirus failed to reduce tumor burden in severe combined immunodeficient mice bearing either B16ova or reovirus-sensitive B16tk metastases. In the human system, reovirus acted solely as an adjuvant when added to dendritic cells already loaded with Mel888, supporting priming of specific antitumor cytotoxic lymphocyte, in the absence of significant direct tumor oncolysis; UV-treated nonreplicating reovirus was similarly immunogenic. CONCLUSION: The immune response is critical in mediating the efficacy of reovirus, and does not depend upon direct viral oncolysis or replication. The findings are of direct relevance to fulfilling the potential of this novel anticancer agent.


Asunto(s)
Citotoxicidad Inmunológica/fisiología , Orthoreovirus Mamífero 3/fisiología , Melanoma Experimental/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Replicación Viral/fisiología , Animales , Línea Celular Tumoral , Células Cultivadas , Humanos , Inmunoterapia Adoptiva/métodos , Orthoreovirus Mamífero 3/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/virología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Virus Oncolíticos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/trasplante , Linfocitos T Citotóxicos/virología , Resultado del Tratamiento , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA