Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 20(9): 1791-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22760544

RESUMO

Treatment of permissive tumors with the oncolytic virus (OV) VSV-Δ51 leads to a robust antitumor T-cell response, which contributes to efficacy; however, many tumors are not permissive to in vivo treatment with VSV-Δ51. In an attempt to channel the immune stimulatory properties of VSV-Δ51 and broaden the scope of tumors that can be treated by an OV, we have developed a potent oncolytic vaccine platform, consisting of tumor cells infected with VSV-Δ51. We demonstrate that prophylactic immunization with this infected cell vaccine (ICV) protected mice from subsequent tumor challenge, and expression of granulocyte-monocyte colony stimulating factor (GM-CSF) by the virus (VSVgm-ICV) increased efficacy. Immunization with VSVgm-ICV in the VSV-resistant B16-F10 model induced maturation of dendritic and natural killer (NK) cell populations. The challenge tumor is rapidly infiltrated by a large number of interferon γ (IFNγ)-producing T and NK cells. Finally, we demonstrate that this approach is robust enough to control the growth of established tumors. This strategy is broadly applicable because of VSV's extremely broad tropism, allowing nearly all cell types to be infected at high multiplicities of infection in vitro, where the virus replication kinetics outpace the cellular IFN response. It is also personalized to the unique tumor antigen(s) displayed by the cancer cell.


Assuntos
Vacinas Anticâncer/imunologia , Melanoma Experimental/prevenção & controle , Melanoma Experimental/terapia , Neoplasias Cutâneas/prevenção & controle , Neoplasias Cutâneas/terapia , Vesiculovirus/imunologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunização , Interferon gama/biossíntese , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Neoplasias Cutâneas/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Vero , Vesiculovirus/genética , Replicação Viral
2.
Int J Dev Biol ; 59(1-3): 141-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26374535

RESUMO

Members of the inhibitor of apoptosis (IAP) family control several critical aspects of innate immunity, cell death, and tumorigenesis. Small molecule antagonists that target specific IAP oncoproteins, primarily cIAP1 and cIAP2, but potentially also XIAP and Livin, modulate distinct immune signal transduction pathways that can lead to an increased sensitivity of tumors cells to cytokine-mediated apoptosis. These antagonists are based on the structure of an endogenous cellular IAP inhibitor called Smac. Smac is normally sequestered within the mitochondria and is released into the cytoplasm upon cell death stimuli, thereby overcoming the anti-apoptotic action of the IAPs. The therapeutic usefulness of recombinant tumoricidal cytokines to treat cancer patients is principally limited due to their unacceptable adverse side effects. Therefore, investigators have sought to develop alternative regimens that do not rely on exogenously delivered death ligands. These approaches include the stimulation of the immune system with oncolytic virus-based agents or Toll-like receptor agonists in combination with Smac mimetics. Similarly, preclinical combination immunotherapy studies reveal that recombinant interferon synergizes with Smac mimetics to kill cancer. This strategy opens up new therapeutic avenues for anti-cancer therapy by modulating specific immune-mediated death pathways employing unique dual-pronged combinatorial approaches.


Assuntos
Apoptose/imunologia , Imunoterapia/métodos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Mitocondriais/metabolismo , Neoplasias/terapia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Proteína 3 com Repetições IAP de Baculovírus , Citocinas/imunologia , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/metabolismo , Interferon alfa-2 , Interferon-alfa/uso terapêutico , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/imunologia , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo
3.
Nat Commun ; 6: 6410, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-25817275

RESUMO

In this study, we show that several microtubule-destabilizing agents used for decades for treatment of cancer and other diseases also sensitize cancer cells to oncolytic rhabdoviruses and improve therapeutic outcomes in resistant murine cancer models. Drug-induced microtubule destabilization leads to superior viral spread in cancer cells by disrupting type I IFN mRNA translation, leading to decreased IFN protein expression and secretion. Furthermore, microtubule-destabilizing agents specifically promote cancer cell death following stimulation by a subset of infection-induced cytokines, thereby increasing viral bystander effects. This study reveals a previously unappreciated role for microtubule structures in the regulation of the innate cellular antiviral response and demonstrates that unexpected combinations of approved chemotherapeutics and biological agents can lead to improved therapeutic outcomes.


Assuntos
Efeito Espectador/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Interferon Tipo I/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Terapia Viral Oncolítica , Vírus Oncolíticos , RNA Mensageiro/efeitos dos fármacos , Infecções por Rhabdoviridae/imunologia , Moduladores de Tubulina/farmacologia , Albendazol/farmacologia , Animais , Benzimidazóis/farmacologia , Efeito Espectador/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Colchicina/farmacologia , Citocinas/imunologia , Células HT29 , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Nocodazol/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Rhabdoviridae , Células Vero , Vimblastina/análogos & derivados , Vimblastina/farmacologia , Vinorelbina
4.
Cancer Cell ; 28(2): 210-24, 2015 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26212250

RESUMO

Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.


Assuntos
Neoplasias/virologia , Vírus Oncolíticos/fisiologia , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/virologia , Humanos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/virologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Interferência de RNA , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Vaccinia virus/fisiologia
5.
Clin Cancer Res ; 19(14): 3832-43, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23714728

RESUMO

PURPOSE: Acute lymphoblastic leukemia (ALL) remains incurable in most adults. It has been difficult to provide effective immunotherapy to improve outcomes for the majority of patients. Rhabdoviruses induce strong antiviral immune responses. We hypothesized that mice administered ex vivo rhabdovirus-infected ALL cells [immunotherapy by leukemia-oncotropic virus (iLOV)] would develop robust antileukemic immune responses capable of controlling ALL. EXPERIMENTAL DESIGN: Viral protein production, replication, and cytopathy were measured in human and murine ALL cells exposed to attenuated rhabdovirus. Survival following injection of graded amounts of ALL cells was compared between cohorts of mice administered γ-irradiated rhabdovirus-infected ALL cells (iLOV) or multiple control vaccines to determine key immunotherapeutic components and characteristics. Host immune requirements were assessed in immunodeficient and bone marrow-transplanted mice or by adoptive splenocyte transfer from immunized donors. Antileukemic immune memory was ascertained by second leukemic challenge in long-term survivors. RESULTS: Human and murine ALL cells were infected and killed by rhabdovirus; this produced a potent antileukemia vaccine. iLOV protected mice from otherwise lethal ALL by developing durable leukemia-specific immune-mediated responses (P < 0.0001), which required an intact CTL compartment. Preexisting antiviral immunity augmented iLOV potency. Splenocytes from iLOV-vaccinated donors protected 60% of naïve recipients from ALL challenge (P = 0.0001). Injecting leukemia cells activated by, or concurrent with, multiple Toll-like receptor agonists could not reproduce the protective effect of iLOV. Similarly, injecting uninfected irradiated viable, apoptotic, or necrotic leukemia cells with/without concurrent rhabdovirus administration was ineffective. CONCLUSION: Rhabdovirus-infected leukemia cells can be used to produce a vaccine that induces robust specific immunity against aggressive leukemia.


Assuntos
Imunoterapia Adotiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Rhabdoviridae/fisiologia , Transferência Adotiva , Animais , Transplante de Medula Óssea , Vacinas Anticâncer , Linhagem Celular Tumoral , Sobrevivência Celular , Chlorocebus aethiops , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Transplante de Neoplasias , Medicina de Precisão , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA