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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Gene Ther ; 23(2): 135-43, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26583803

RESUMEN

Although most high-risk neuroblastomas are responsive to chemotherapy, relapse is common and long-term survival is < 40%, underscoring the need for more effective treatments. We evaluated the responsiveness of 12 neuroblastoma cell lines to the Δγ134.5 attenuated oncolytic herpes simplex virus (oHSV), Seprehvir (HSV1716), which is currently used in pediatric phase I trials. We found that entry of Seprehvir in neuroblastoma cells is independent of the expression of nectin-1 and the sum of all four known major HSV entry receptors. We observed varying levels of sensitivity and permissivity to Seprehvir, suggesting that the cellular anti-viral response, not virus entry, is the key determinant of efficacy with this virus. In vivo, we found significant anti-tumor efficacy following Seprehvir treatment, which ranged from 6/10 complete responses in the CHP-134 model to a mild prolonged median survival in the SK-N-AS model. Taken together, these data suggest that anti-tumor efficacy cannot be solely predicted based on in vitro response. Whether or not this discordance holds true for other viruses or tumor types is unknown. Our results also suggest that profiling the expression of known viral entry receptors on neuroblastoma cells may not be entirely predictive of their susceptibility to Seprehvir therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Herpesvirus Humano 1 , Neuroblastoma/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Receptores Virales/metabolismo , Internalización del Virus , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Ratones , Ratones Desnudos , Neuroblastoma/inmunología , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Br J Cancer ; 101(5): 774-81, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19707204

RESUMEN

BACKGROUND: Interruption of the role of p53s as a tumour suppressor by MDM2 may be one of the mechanisms by which cancer cells evade current therapy. Blocking the inhibition of wild-type p53 by MDM2 in cancer cells should reactivate p53's tumour suppressor functions and enhance current cancer treatments. MI-63 is a novel non-peptide small molecule that has shown strong binding affinity (K(i)=3 nM) for MDM2; however, its effects on paediatric cancer cells and the specific mechanism of tumour suppressor reactivation have not been evaluated. METHODS: Rhabdomyosarcoma (RMS), the most common childhood soft tissue sarcoma, expresses either wild-type or mutant p53 protein. We examined the inhibitory effects of MI-63 in embryonal RMS (ERMS) and alveolar RMS (ARMS) cell lines expressing wild-type or mutated p53. RESULTS: Treatment with MI-63 reduced cell viability by 13.4% and by <1%, respectively, at 72 h in both RH36 and RH18 cell lines expressing wild-type p53. In contrast, RH30 and RD2 cells expressing p53 mutants are resistant to MI-63 treatment. An increased expression of p53, p21(WAF1), and Bax protein was observed after treatment with MI-63 in RMS cells with wild-type p53, and apoptosis was confirmed by cleaved PARP and caspase-3 expression. However, RD2 and RH30 RMS cells, as well as human normal skeletal muscle cells, showed a minimal increase in p53 signalling and no induction of cleaved PARP and caspase-3. MI-63 was compared with Nutlin-3, a known MDM2 inhibitor, and was found to be more potent in the inhibition of cell proliferation/viability. Further, synergy was observed when MI-63 was used in combination with doxorubicin. CONCLUSION: These results indicate that MI-63 is a potent therapeutic agent for RMS cells expressing wild-type p53 protein.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Indoles/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Rabdomiosarcoma Alveolar/patología , Rabdomiosarcoma Embrionario/patología , Compuestos de Espiro/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/química , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Imidazoles/farmacología , Indoles/química , Peso Molecular , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Rabdomiosarcoma Alveolar/tratamiento farmacológico , Rabdomiosarcoma Alveolar/metabolismo , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Rabdomiosarcoma Embrionario/metabolismo , Transducción de Señal/efectos de los fármacos , Compuestos de Espiro/química , Relación Estructura-Actividad , Proteína p53 Supresora de Tumor/efectos de los fármacos
3.
Br J Cancer ; 100(1): 106-12, 2009 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-19127268

RESUMEN

Persistent activation of the signal transducer and activator of transcription 3 (STAT3) signalling has been linked to oncogenesis and the development of chemotherapy resistance in glioblastoma and other cancers. Inhibition of the STAT3 pathway thus represents an attractive therapeutic approach for cancer. In this study, we investigated the inhibitory effects of a small molecule compound known as LLL-3, which is a structural analogue of the earlier reported STAT3 inhibitor, STA-21, on the cell viability of human glioblastoma cells, U87, U373, and U251 expressing constitutively activated STAT3. We also investigated the inhibitory effects of LLL-3 on U87 glioblastoma cell growth in a mouse tumour model as well as the impact it had on the survival time of the treated mice. We observed that LLL-3 inhibited STAT3-dependent transcriptional and DNA binding activities. LLL-3 also inhibited viability of U87, U373, and U251 glioblastoma cells as well as induced apoptosis of these glioblastoma cell lines as evidenced by increased poly (ADP-ribose) polymerase (PARP) and caspase-3 cleavages. Furthermore, the U87 glioblastoma tumour-bearing mice treated with LLL-3 exhibited prolonged survival relative to vehicle-treated mice (28.5 vs 16 days) and had smaller intracranial tumours and no evidence of contralateral invasion. These results suggest that LLL-3 may be a potential therapeutic agent in the treatment of glioblastoma with constitutive STAT3 activation.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , ADN/metabolismo , Modelos Animales de Enfermedad , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factor de Transcripción STAT3/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA