Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 21(3): 561-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23070115

RESUMO

Glioblastoma multiforme (GBM) remains an untreatable human brain malignancy. Despite promising preclinical studies using oncolytic herpes simplex virus (oHSV) vectors, efficacy in patients has been limited by inefficient virus replication in tumor cells. This disappointing outcome can be attributed in part to attenuating mutations engineered into these viruses to prevent replication in normal cells. Alternatively, retargeting of fully replication-competent HSV to tumor-associated receptors has the potential to achieve tumor specificity without impairment of oncolytic activity. Here, we report the establishment of an HSV retargeting system that relies on the combination of two engineered viral glycoproteins, gD and gB, to mediate highly efficient HSV infection exclusively through recognition of the abundantly expressed epidermal growth factor receptor (EGFR) on glioblastoma cells. We demonstrate efficacy in vitro and in a heterotopic tumor model in mice. Evidence for systemically administered virus homing to the tumor mass is presented. Treatment of orthotopic primary human GBM xenografts demonstrated prolonged survival with up to 73% of animals showing a complete response as confirmed by magnetic resonance imaging. Our study describes an approach to HSV retargeting that is effective in a glioma model and may be applicable to the treatment of a broad range of tumor types.


Assuntos
Receptores ErbB/metabolismo , Glioblastoma/terapia , Terapia Viral Oncolítica/métodos , Simplexvirus/genética , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetinae , Feminino , Vetores Genéticos , Células HT29 , Humanos , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Plasmídeos , Recombinação Genética , Simplexvirus/fisiologia , Resultado do Tratamento , Células Vero , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Mol Ther ; 21(5): 1014-23, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23481323

RESUMO

Understanding the host response to oncolytic viruses is important to maximize their antitumor efficacy. Despite robust cytotoxicity and high virus production of an oncolytic herpes simplex virus (oHSV) in cultured human sarcoma cells, intratumoral (ITu) virus injection resulted in only mild antitumor effects in some xenograft models, prompting us to characterize the host inflammatory response. Virotherapy induced an acute neutrophilic infiltrate, a relative decrease of ITu macrophages, and a myeloid cell-dependent upregulation of host-derived vascular endothelial growth factor (VEGF). Anti-VEGF antibodies, bevacizumab and r84, the latter of which binds VEGF and selectively inhibits binding to VEGF receptor-2 (VEGFR2) but not VEGFR1, enhanced the antitumor effects of virotherapy, in part due to decreased angiogenesis but not increased virus production. Neither antibody affected neutrophilic infiltration but both partially mitigated virus-induced depletion of macrophages. Enhancement of virotherapy-mediated antitumor effects by anti-VEGF antibodies could largely be recapitulated by systemic depletion of CD11b(+) cells. These data suggest the combined effect of oHSV virotherapy and anti-VEGF antibodies is in part due to modulation of a host inflammatory reaction to virus. Our data provide strong preclinical support for combined oHSV and anti-VEGF antibody therapy and suggest that understanding and counteracting the innate host response may help enable the full antitumor potential of oncolytic virotherapy.


Assuntos
Vetores Genéticos/imunologia , Células Mieloides/imunologia , Neoplasias/imunologia , Vírus Oncolíticos/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacologia , Bevacizumab , Antígeno CD11b/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Células Mieloides/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Neovascularização Patológica/terapia , Terapia Viral Oncolítica , Sarcoma/imunologia , Sarcoma/metabolismo , Sarcoma/terapia , Simplexvirus/imunologia , Células Estromais/metabolismo , Células Estromais/virologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/imunologia , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Ther ; 20(2): 287-97, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22031239

RESUMO

Here, we describe the construction and testing of a novel herpes simplex virus type 1 (HSV-1) derived oncolytic virus (OV): 34.5ENVE (viral ICP34.5 Expressed by Nestin promotor and Vstat120 Expressing), for the treatment of cancer. This virus showed significant glioma-specific killing and antiangiogenic effects in vitro and in vivo. Treatment of subcutaneous and intracranial glioma-bearing mice with 34.5ENVE showed a significant increase in median survival of mice in four different glioma models. Histology and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) revealed reduced microvessel density (MVD) and increased tumoral necrosis in 34.5ENVE-treated tumor tissue compared to control OV-treated tumor tissue. Collectively, these results describe the construction, efficacy, and impact on tumor microenvironment of a transcriptionally driven OV armed with Vstat120 gene expression. These preclinical results will facilitate future clinical testing of 34.5ENVE.


Assuntos
Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Linhagem Celular , Movimento Celular , Efeito Citopatogênico Viral , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Feminino , Expressão Gênica , Ordem dos Genes , Terapia Genética , Glioma/genética , Glioma/terapia , Humanos , Proteínas de Filamentos Intermediários/genética , Camundongos , Camundongos Nus , Necrose/genética , Neoplasias/genética , Neoplasias/mortalidade , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Proteínas do Tecido Nervoso/genética , Nestina , Análise de Sobrevida , Replicação Viral
4.
Cancer Res ; 74(6): 1752-65, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24453002

RESUMO

Glioblastoma is the most common and aggressive histologic subtype of brain cancer with poor outcomes and limited treatment options. Here, we report the selective overexpression of the protein arginine methyltransferase PRMT5 as a novel candidate theranostic target in this disease. PRMT5 silences the transcription of regulatory genes by catalyzing symmetric dimethylation of arginine residues on histone tails. PRMT5 overexpression in patient-derived primary tumors and cell lines correlated with cell line growth rate and inversely with overall patient survival. Genetic attenuation of PRMT5 led to cell-cycle arrest, apoptosis, and loss of cell migratory activity. Cell death was p53-independent but caspase-dependent and enhanced with temozolomide, a chemotherapeutic agent used as a present standard of care. Global gene profiling and chromatin immunoprecipitation identified the tumor suppressor ST7 as a key gene silenced by PRMT5. Diminished ST7 expression was associated with reduced patient survival. PRMT5 attenuation limited PRMT5 recruitment to the ST7 promoter, led to restored expression of ST7 and cell growth inhibition. Finally, PRMT5 attenuation enhanced glioblastoma cell survival in a mouse xenograft model of aggressive glioblastoma. Together, our findings defined PRMT5 as a candidate prognostic factor and therapeutic target in glioblastoma, offering a preclinical justification for targeting PRMT5-driven oncogenic pathways in this deadly disease.


Assuntos
Neoplasias Encefálicas/enzimologia , Glioblastoma/enzimologia , Proteína-Arginina N-Metiltransferases/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Proliferação de Células , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/mortalidade , Glioblastoma/terapia , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Knockout , Camundongos Nus , Terapia de Alvo Molecular , Transplante de Neoplasias , Proteína-Arginina N-Metiltransferases/metabolismo , RNA Interferente Pequeno/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/metabolismo
5.
PLoS One ; 8(8): e71932, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936533

RESUMO

Recent studies report that STAT3 signaling is a master regulator of mesenchymal transformation of gliomas and that STAT3 modulated genes are highly expressed in the mesenchymal transcriptome of gliomas. A currently studied experimental treatment for gliomas consists of intratumoral injection of oncolytic viruses (OV), such as oncolytic herpes simplex virus type 1 (oHSV). We have described one particular oHSV (rQNestin34.5) that exhibits potent anti-glioma activity in animal models. Here, we hypothesized that alterations in STAT3 signaling in glioma cells may affect the replicative ability of rQNestin34.5. In fact, human U251 glioma cells engineered to either over-express STAT3 or with genetic down-regulation of STAT3 supported oHSV replication to a significantly higher or lesser degree, respectively, when compared to controls. Administration of pharmacologic agents that increase STAT3 phosphorylation/activation (Valproic Acid) or increase STAT3 levels (Interleukin 6) also significantly enhanced oHSV replication. Instead, administration of inhibitors of STAT3 phosphorylation/activation (LLL12) significantly reduced oHSV replication. STAT3 led to a reduction in interferon signaling in oHSV infected cells and inhibition of interferon signaling abolished the effect of STAT3 on oHSV replication. These data thus indicate that STAT3 signaling in malignant gliomas enhances oHSV replication, likely by inhibiting the interferon response in infected glioma cells, thus suggesting avenues for possible potentiation of oncolytic virotherapy.


Assuntos
Neoplasias Encefálicas/virologia , Glioma/virologia , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Terapia Viral Oncolítica , Fator de Transcrição STAT3/metabolismo , Replicação Viral , Anticonvulsivantes/farmacologia , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Proliferação de Células , Terapia Combinada , Glioma/metabolismo , Glioma/terapia , Herpes Simples/genética , Herpes Simples/terapia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Luciferases/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Células Tumorais Cultivadas , Ácido Valproico/farmacologia
6.
Clin Cancer Res ; 19(21): 5952-9, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24056786

RESUMO

PURPOSE: Oncolytic viruses (OV) based on herpes simplex virus type 1 (HSV1) are being used in clinical trials for a variety of cancers. The OV, rQNestin34.5, uses a nestin promoter/enhancer to selectively drive robust viral replication in malignant glioma cells. We have discovered that this promoter becomes extensively methylated in infected glioma cells, reducing OV efficacy. EXPERIMENTAL DESIGN: We used demethylating drugs [5-azacytidine (5-Aza)], decitabine, or valproic acid (VPA) in both in vitro and in vivo malignant glioma models to determine if they improved the efficacy of rQNestin34.5 therapy. RESULTS: The use of demethylating agents, such as 5-Aza, improved OV replication and tumor cell lysis in vitro and, in fact, synergized pharmacologically on Chou-Talalay analysis. In vivo, the combination of the demethylating agents, 5-Aza or decitabine, with rQNestin34.5 significantly prolonged the survivorship of athymic mice harboring intracranial human glioma xenografts over single agent alone. CONCLUSION: These results, thus, provide further justification for the exploration of demethylating agents when combined with the OV, rQNestin34.5, in preclinical therapeutics and, possibly, clinical trials for malignant glioma.


Assuntos
Azacitidina/farmacologia , Metilação de DNA/efeitos dos fármacos , Vetores Genéticos/genética , Glioma/genética , Herpesvirus Humano 1/genética , Vírus Oncolíticos/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ilhas de CpG , Modelos Animais de Doenças , Elementos Facilitadores Genéticos , Feminino , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Vetores Genéticos/administração & dosagem , Glioma/mortalidade , Glioma/patologia , Glioma/terapia , Humanos , Camundongos , Nestina/genética , Terapia Viral Oncolítica , Regiões Promotoras Genéticas , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Res ; 72(6): 1353-62, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22282654

RESUMO

Oncolytic viral therapy has been explored widely as an option for glioma treatment but its effectiveness has remained limited. Cysteine rich 61 (CCN1) is an extracellular matrix (ECM) protein elevated in cancer cells that modulates their adhesion and migration by binding cell surface receptors. In this study, we examined a hypothesized role for CCN1 in limiting the efficacy of oncolytic viral therapy for glioma, based on evidence of CCN1 induction that occurs in this setting. Strikingly, we found that exogenous CCN1 in glioma ECM orchestrated a cellular antiviral response that reduced viral replication and limited cytolytic efficacy. Gene expression profiling and real-time PCR analysis revealed a significant induction of type-I interferon responsive genes in response to CCN1 exposure. This induction was accompanied by activation of the Jak/Stat signaling pathway, consistent with induction of an innate antiviral cellular response. Both effects were mediated by the binding of CCN1 to the cell surface integrin α6ß1, activating its signaling and leading to rapid secretion of interferon-α, which was essential for the innate antiviral effect. Together, our findings reveal how an integrin signaling pathway mediates activation of a type-I antiviral interferon response that can limit the efficacy of oncolytic viral therapy. Furthermore, they suggest therapeutic interventions to inhibit CCN1-integrin α6 interactions to sensitize gliomas to viral oncolysis.


Assuntos
Neoplasias Encefálicas/terapia , Proteína Rica em Cisteína 61/imunologia , Glioma/terapia , Terapia Viral Oncolítica , Animais , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Humanos , Integrina alfa6beta1/imunologia , Interferon Tipo I/imunologia , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Camundongos , Camundongos Nus , Transdução de Sinais
8.
Clin Cancer Res ; 18(18): 4931-41, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22753591

RESUMO

PURPOSE: Copper in serum supports angiogenesis and inhibits replication of wild-type HSV-1. Copper chelation is currently being investigated as an antiangiogenic and antineoplastic agent in patients diagnosed with cancer. Herpes simplex virus-derived oncolytic viruses (oHSV) are being evaluated for safety and efficacy in patients, but several host barriers limit their efficacy. Here, we tested whether copper inhibits oHSV infection and replication and whether copper chelation would augment therapeutic efficacy of oHSV. EXPERIMENTAL DESIGN: Subcutaneous and intracranial tumor-bearing mice were treated with oHSV ± ATN-224 to evaluate tumor burden and survival. Virus replication and cell killing was measured in the presence or absence of the copper chelating agent ATN-224 and in the presence or absence of copper in vitro. Microvessel density and changes in perfusion were evaluated by immunohistochemistry and dynamic contrast enhanced MRI (DCE-MRI). Serum stability of oHSV was measured in mice fed with ATN-224. Tumor-bearing mice were injected intravenously with oHSV; tumor burden and amount of virus in tumor tissue were evaluated. RESULTS: Combination of systemic ATN-224 and oHSV significantly reduced tumor growth and prolonged animal survival. Immunohistochemistry and DCE-MRI imaging confirmed that ATN-224 reduced oHSV-induced blood vessel density and vascular leakage. Copper at physiologically relevant concentrations inhibited oHSV replication and glioma cell killing, and this effect was rescued by ATN-224. ATN-224 increased serum stability of oHSV and enhanced the efficacy of systemic delivery. CONCLUSION: This study shows that combining ATN-224 with oHSV significantly increased serum stability of oHSV and greatly enhanced its replication and antitumor efficacy.


Assuntos
Antineoplásicos/uso terapêutico , Quelantes/uso terapêutico , Cobre , Molibdênio/uso terapêutico , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Simplexvirus/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Quelantes/farmacologia , Terapia Combinada , Efeito Citopatogênico Viral/efeitos dos fármacos , Feminino , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glioma/diagnóstico , Glioma/genética , Glioma/mortalidade , Glioma/terapia , Humanos , Camundongos , Camundongos Nus , Molibdênio/farmacologia , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/mortalidade , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Recent Pat CNS Drug Discov ; 4(1): 1-13, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19149710

RESUMO

The World Health Organization grossly classifies the various types of astrocytomas using a grade system with grade IV gliomas having the worst prognosis. Oncolytic virus therapy is a novel treatment option for GBM patients. Several patents describe various oncolytic viruses used in preclinical and clinical trials to evaluate safety and efficacy. These viruses are natural or genetically engineered from different viruses such as HSV-1, Adenovirus, Reovirus, and New Castle Disease Virus. While several anecdotal studies have indicated therapeutic advantage, recent clinical trials have revealed the safety of their usage, but demonstration of significant efficacy remains to be established. Oncolytic viruses are being redesigned with an interest in combating the tumor microenvironment in addition to defeating the cancerous cells. Several patents describe the inclusion of tumor microenvironment modulating genes within the viral backbone and in particular those which attack the tumor angiotome. The very innovative approaches being used to improve therapeutic efficacy include: design of viruses which can express cytokines to activate a systemic antitumor immune response, inclusion of angiostatic genes to combat tumor vasculature, and also enzymes capable of digesting tumor extra cellular matrix (ECM) to enhance viral spread through solid tumors. As increasingly more novel viruses are being tested and patented, the future battle against glioma looks promising.


Assuntos
Glioma/terapia , Terapia Viral Oncolítica/métodos , Animais , Humanos , Terapia Viral Oncolítica/tendências
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA