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










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2058: 295-306, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31486047

RESUMO

Fluorescence in situ hybridization (FISH) is a specific, sensitive, accurate, and reliable technique widely applied in both research and clinic. Here we describe the detailed protocol of a FISH method established by us to serve the scientific purposes of the first oncolytic parvovirus clinical trial (ParvOryx01). This trial was launched in Germany in 2011. After trial completion in 2015, results were published in Molecular Therapy in 2017. The primary purpose of the trial was to evaluate the safety of an oncolytic parvovirus, H-1PV (ParvOryx), in recurrent glioblastoma patients. In addition, the efficiency of H-1PV tumor targeting after intratumoral or systemic virus administration was assessed by FISH detection of viral nucleic acids (genomic single-stranded DNA, mRNA and parvovirus double-stranded replicative forms) in formalin-fixed paraffin-embedded glioblastoma tissues resected at day 10 after ParvOryx treatment. The FISH method allowed the detection-for the first time in humans-of H-1PV replication markers in brain tumors of parvovirus-treated patients. A protocol combining mRNA FISH with simultaneous immunofluorescent staining for tumor and tumor microenvironment markers was also developed and is described here, in order to better characterize H-1PV cellular targets and H-1PV treatment-associated tumor microenvironment changes.


Assuntos
Neoplasias Encefálicas/diagnóstico , DNA Viral , Vetores Genéticos , Parvovirus H-1 , Hibridização in Situ Fluorescente , Vírus Oncolíticos , Neoplasias Encefálicas/terapia , Imunofluorescência , Vetores Genéticos/genética , Parvovirus H-1/genética , Parvovirus H-1/imunologia , Humanos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Microambiente Tumoral , Replicação Viral
2.
Mol Ther ; 25(12): 2620-2634, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-28967558

RESUMO

Oncolytic virotherapy may be a means of improving the dismal prognosis of malignant brain tumors. The rat H-1 parvovirus (H-1PV) suppresses tumors in preclinical glioma models, through both direct oncolysis and stimulation of anticancer immune responses. This was the basis of ParvOryx01, the first phase I/IIa clinical trial of an oncolytic parvovirus in recurrent glioblastoma patients. H-1PV (escalating dose) was administered via intratumoral or intravenous injection. Tumors were resected 9 days after treatment, and virus was re-administered around the resection cavity. Primary endpoints were safety and tolerability, virus distribution, and maximum tolerated dose (MTD). Progression-free and overall survival and levels of viral and immunological markers in the tumor and peripheral blood were also investigated. H-1PV treatment was safe and well tolerated, and no MTD was reached. The virus could cross the blood-brain/tumor barrier and spread widely through the tumor. It showed favorable pharmacokinetics, induced antibody formation in a dose-dependent manner, and triggered specific T cell responses. Markers of virus replication, microglia/macrophage activation, and cytotoxic T cell infiltration were detected in infected tumors, suggesting that H-1PV may trigger an immunogenic stimulus. Median survival was extended in comparison with recent meta-analyses. Altogether, ParvOryx01 results provide an impetus for further H-1PV clinical development.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/terapia , Parvovirus H-1/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Feminino , Expressão Gênica , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Radioterapia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Transgenes , Resultado do Tratamento
3.
Neoplasia ; 17(7): 564-73, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26297434

RESUMO

The oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) is overactivated in malignant glioma and plays a key role in promoting cell survival, thereby increasing the acquired apoptosis resistance of these tumors. Here we investigated the STAT3/myeloid cell leukemia 1 (MCL1) signaling pathway as a target to overcome the resistance of glioma cells to the Bcl-2-inhibiting synthetic BH3 mimetic ABT-737. Stable lentiviral knockdown of MCL1 sensitized LN229 and U87 glioma cells to apoptotic cell death induced by single-agent treatment with ABT-737 which was associated with an early activation of DEVDase activity, cytochrome c release, and nuclear apoptosis. Similar sensitizing effects were observed when ABT-737 treatment was combined with the multikinase inhibitor sorafenib which effectively suppressed levels of phosphorylated STAT3 and MCL1 in MCL1-proficient LN229 and U87 glioma cells. In analogous fashion, these synergistic effects were observed when we combined ABT-737 with the STAT3 inhibitor WP-1066. Lentiviral knockdown of the activating transcription factor 5 combined with subsequent quantitative polymerase chain reaction analysis revealed that sorafenib-dependent suppression of MCL1 occurred at the transcriptional level but did not depend on activating transcription factor 5 which previously had been proposed to be essential for MCL1-dependent glioma cell survival. In contrast, the constitutively active STAT3 mutant STAT3-C was able to significantly enhance MCL1 levels under sorafenib treatment to retain cell survival. Collectively, these data demonstrate that sorafenib targets MCL1 in a STAT3-dependent manner, thereby sensitizing glioma cells to treatment with ABT-737. They also suggest that targeting STAT3 in combination with inducers of the intrinsic pathway of apoptosis may be a promising novel strategy for the treatment of malignant glioma.


Assuntos
Compostos de Bifenilo/farmacologia , Glioma/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Niacinamida/análogos & derivados , Nitrofenóis/farmacologia , Compostos de Fenilureia/farmacologia , Fator de Transcrição STAT3/metabolismo , Sulfonamidas/farmacologia , Fatores Ativadores da Transcrição/genética , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Citocromos c/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Niacinamida/farmacologia , Peptídeo Hidrolases/metabolismo , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Piridinas/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Sorafenibe , Tirfostinas/farmacologia
4.
Curr Opin Virol ; 13: 17-24, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25841215

RESUMO

The H-1 parvovirus (H-1PV) exerts oncosuppressive action that has two components: oncotoxicity and immunostimulation. While many human tumor cells, including conventional drug-resistant ones, can be killed by H-1PV, some fail to support progeny virus production, necessary for infection propagation in neoplastic tissues. This limitation can be overcome through forced selection of H-1PV variants capable of enhanced multiplication and spreading in human tumor cells. In the context of further developing H-1PV for use in cancer therapy, arming it with immunostimulatory CpG motifs under conditions preserving replication and oncolysis enhances its action as an anticancer vaccine adjuvant. A first clinical study of H-1PV treatment in glioma patients has yielded evidence of intratumoral synthesis of the viral oncotoxic protein NS1 and immune cell infiltration.


Assuntos
Alphaherpesvirinae/fisiologia , Parvovirus H-1/fisiologia , Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Alphaherpesvirinae/genética , Animais , Parvovirus H-1/genética , Humanos , Neoplasias/imunologia , Neoplasias/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética
5.
Clin Cancer Res ; 17(16): 5333-42, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21715567

RESUMO

PURPOSE: In previous studies, we have shown that the apathogenic rat parvovirus H-1 (H-1PV) is capable to induce regression of advanced symptomatic rat and human gliomas in a rat model, when the virus was injected in the tumor (intracranially) or intravenously. Infection with H-1PV did not provoke any pathology in nontumor tissue. This study addresses the question whether also intranasal application of this oncolytic virus is suitable and sufficient for treating gliomas in this animal model. EXPERIMENTAL DESIGN: Rat (RG-2) or human (U87) glioma cells were grafted stereotactically in the brain of rats (Wistar or RNU, respectively), and after development of tumors visible by MRI, H-1PV was instilled intranasally. Tumor regression was monitored by MRI, and survival was analyzed by Kaplan-Meier analysis. Brains from sacrificed animals were analyzed for histologic alterations, presence of viral DNA and proteins and infectious virions. In addition, distribution of virus to other organs was determined. RESULTS: A single intranasal instillation of H-1PV was sufficient to induce efficient regression of rat glioma, leading to significant prolongation of survival without any toxicity for other tissues. It is shown that the virus reaches brain and other tissues, and that the viral replication-associated (and oncolysis-associated) regulatory proteins are exclusively expressed in the tumor tissue. In rats with xenografts of human glioma, oncolytic activity of H-1PV was less pronounced, however, leading to significant prolongation of survival. CONCLUSION: In view of an ongoing clinical trial on the use of H-1PV for oncolytic virotherapy of glioma, the option of applying the virus intranasally may be a valuable alternative to invasive routes of infection.


Assuntos
Glioma/terapia , Parvovirus H-1/fisiologia , Terapia Viral Oncolítica/métodos , Infecções por Parvoviridae/terapia , Administração Intranasal , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular Tumoral , DNA Viral/análise , Modelos Animais de Doenças , Feminino , Glioma/patologia , Glioma/virologia , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Resultado do Tratamento , Carga Tumoral , Proteínas Virais/análise , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cytokine Growth Factor Rev ; 21(2-3): 185-95, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20211577

RESUMO

The experimental infectivity and excellent tolerance of some rodent autonomous parvoviruses in humans, together with their oncosuppressive effects in preclinical models, speak for the inclusion of these agents in the arsenal of oncolytic viruses under consideration for cancer therapy. In particular, wild-type parvovirus H-1PV can achieve a complete cure of various tumors in animal models and kill tumor cells that resist conventional anticancer treatments. There is growing evidence that H-1PV oncosuppression involves an immune component in addition to the direct viral oncolytic effect. This article summarizes the recent assessment of H-1PV antineoplastic activity in glioma, pancreatic ductal adenocarcinoma, and non-Hodgkin lymphoma models, laying the foundation for the present launch of a first phase I/IIa clinical trial on glioma patients.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Parvovirus , Animais , Ensaios Clínicos como Assunto , Humanos , Fatores Imunológicos/metabolismo , Neoplasias/patologia , Neoplasias/virologia , Parvovirus/genética , Parvovirus/fisiologia
7.
Neuro Oncol ; 12(8): 804-14, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20299703

RESUMO

Oncolytic virotherapy is a potential treatment modality under investigation for various malignancies including malignant brain tumors. Unlike some other natural or modified viruses that show oncolytic activity against cerebral neoplasms, the rodent parvovirus H-1 (H-1PV) is completely apathogenic in humans. H-1PV efficiently kills a number of tumor cells without harm to corresponding normal ones. In this study, the concept of H-1PV-based virotherapy of glioma was tested for rat (RG-2 cell-derived) and for human (U87 cell-derived) gliomas in immunocompetent and immunodeficient rat models, respectively. Large orthotopic rat and human glioma cell-derived tumors were treated with either single stereotactic intratumoral or multiple intravenous (iv) H-1PV injections. Oncolysis was monitored by magnetic resonance imaging and proven by histology. Virus distribution and replication were determined in brain and organs. In immunocompetent rats bearing RG-2-derived tumors, a single stereotactic intratumoral injection of H-1PV and multiple systemic (iv) applications of the virus were sufficient for remission of advanced and even symptomatic intracranial gliomas without damaging normal brain tissue or other organs. H-1PV therapy resulted in significantly improved survival (Kaplan-Meier analysis) in both the rat and human glioma models. Virus replication in tumors indicated a contribution of secondary infection by progeny virus to the efficiency of oncolysis. Virus replication was restricted to tumors, although H-1PV DNA could be detected transiently in adjacent or remote normal brain tissue and in noncerebral tissues. The results presented here and the innocuousness of H-1PV for humans argue for the use of H-1PV as a powerful means to perform oncolytic therapy of malignant gliomas.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Terapia Viral Oncolítica/métodos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Encéfalo/patologia , Encéfalo/virologia , Neoplasias Encefálicas/patologia , DNA Viral/isolamento & purificação , Modelos Animais de Doenças , Glioma/patologia , Parvovirus H-1 , Humanos , Imageamento por Ressonância Magnética , Reação em Cadeia da Polimerase , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Ther ; 17(7): 1164-72, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19367260

RESUMO

The incidence of lymphomas developing in both immunocompetent and immunosuppressed patients continues to steadily increase worldwide. Current chemotherapy and immunotherapy approaches have several limitations, such as severe side toxicity and selection of resistant cell variants. Autonomous parvoviruses (PVs), in particular the rat parvovirus H-1PV, have emerged as promising anticancer agents. Although it is apathogenic in humans, H-1PV has been shown to infect and suppress various rat and human tumors in animal models. In this study, we demonstrate the capacity of H-1PV for efficiently killing, through necrosis, cell cultures originating from Burkitt's lymphoma (BL), while sparing normal B lymphocytes. The cytotoxic effect was generally accompanied by a productive H-1PV infection. Remarkably, parvovirus-based monotherapy efficiently suppressed established BL at an advanced stage in a severe combined immunodeficient (SCID) mouse model of the disease. The data show for the first time that an oncolytic parvovirus deserves further consideration as a potential tool for the treatment of some non-Hodgkin B-cell lymphomas, including those resistant to apoptosis induction by rituximab.


Assuntos
Linfoma/terapia , Terapia Viral Oncolítica/métodos , Parvovirus/fisiologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Linfoma de Células B/terapia , Camundongos , Camundongos SCID , Necrose/virologia , Parvovirus/genética , Ratos , Replicação Viral/genética
9.
Exp Neurol ; 189(2): 252-60, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15380477

RESUMO

FGF-2, a potent multifunctional and neurotrophic growth factor, is widely expressed in the brain and upregulated in cerebral ischemia. Previous studies have shown that intraventricularly or systemically administered FGF-2 reduces the size of cerebral infarcts. Whether endogenous FGF-2 is beneficial for the outcome of cerebral ischemia has not been investigated. We have used mice with a null mutation of the fgf2 gene to explore the relevance of endogenous FGF-2 in brain ischemia. Focal cerebral ischemia was produced by occlusion of the middle cerebral artery (MCAO). We found a 75% increase in infarct volume in fgf2 knock-out mice versus wild type littermates (P < 0.05). This difference in the extent of ischemic damage was observed after 24 h, and correlated with decreased viability in fgf2 mutant mice following MCA occlusion. Increased infarct volume in fgf2 null mice was associated with a loss of induction in hippocampal BDNF and trkB mRNA expression. These findings indicate that signaling through trkB may contribute to ameliorating brain damage following ischemia and that bdnf and trkB may be target genes of FGF-2. Together, our data provide the first evidence that endogenous FGF-2 is important in coping with ischemic brain damage suggesting fgf2 as one crucial target gene for new therapeutic strategies in brain ischemia.


Assuntos
Infarto Encefálico/genética , Isquemia Encefálica/genética , Fator Neurotrófico Derivado do Encéfalo/genética , Fator 2 de Crescimento de Fibroblastos/deficiência , Hipocampo/metabolismo , RNA Mensageiro/metabolismo , Animais , Infarto Encefálico/metabolismo , Infarto Encefálico/fisiopatologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo/genética , Fator 2 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/genética , Hipocampo/patologia , Hipocampo/fisiopatologia , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Camundongos , Camundongos Knockout , Receptor trkB/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...