Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
FEBS Lett ; 588(5): 653-8, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24440350

RESUMEN

The MET receptor tyrosine kinase is deregulated primarily via overexpression or point mutations in various human cancers and different strategies for MET inhibition are currently evaluated in clinical trials. We observed by Western blot analysis and by Flow cytometry that MET inhibition by different MET small molecule inhibitors surprisingly increases in a dose-dependent manner total MET levels in treated cells. Mechanistically, this inhibition-related MET accumulation was associated with reduced Tyr1003 phosphorylation and MET physical association with the CBL ubiquitin ligase with concomitant decrease in MET ubiquitination. These data may suggest careful consideration for design of anti-MET clinical protocols.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Indoles/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Sulfonamidas/farmacología , Sulfonas/farmacología , Animales , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Crizotinib , Humanos , Lisosomas/metabolismo , Ratones , Mutación Missense , Células 3T3 NIH , Proteolisis , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Pirazoles/farmacología , Piridazinas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Ubiquitinación
2.
Mol Cancer Ther ; 12(11): 2415-24, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24061647

RESUMEN

The receptor tyrosine kinase MET is a prime target in clinical oncology due to its aberrant activation and involvement in the pathogenesis of a broad spectrum of malignancies. Similar to other targeted kinases, primary and secondary mutations seem to represent an important resistance mechanism to MET inhibitors. Here, we report the biologic activity of a novel MET inhibitor, EMD1214063, on cells that ectopically express the mutated MET variants M1268T, Y1248H, H1112Y, L1213V, H1112L, V1110I, V1206L, and V1238I. Our results show a dose-dependent decrease in MET autophosphorylation in response to EMD1214063 in five of the eight cell lines (IC50 2-43 nmol/L). Blockade of MET by EMD1214063 was accompanied by a reduced activation of downstream effectors in cells expressing EMD1214063-sensitive mutants. In all sensitive mutant-expressing lines, EMD1214063 altered cell-cycle distribution, primarily with an increase in G1 phase. EMD1214063 strongly influenced MET-driven biologic functions, such as cellular morphology, MET-dependent cell motility, and anchorage-independent growth. To assess the in vivo efficacy of EMD1214063, we used a xenograft tumor model in immunocompromised mice bearing NIH3T3 cells expressing sensitive and resistant MET-mutated variants. Animals were randomized for the treatment with EMD1214063 (50 mg/kg/d) or vehicle only. Remarkably, five days of EMD1214063 treatment resulted in a complete regression of the sensitive H1112L-derived tumors, whereas tumor growth remained unaffected in mice with L1213V tumors and in vehicle-treated animals. Collectively, the current data identifies EMD1214063 as a potent MET small-molecule inhibitor with selective activity towards mutated MET variants.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Piridazinas/farmacología , Pirimidinas/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Femenino , Variación Genética , Humanos , Ratones , Células 3T3 NIH , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Mutación Puntual , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-met/metabolismo , Piridazinas/administración & dosificación , Pirimidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Lett ; 289(2): 228-36, 2010 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-19783361

RESUMEN

Point mutations emerge as one of the rate-limiting steps in tumor response to small molecule inhibitors of protein kinases. Here we characterized the response of the MET mutated variants, V1110I, V1238I, V1206L and H1112L to the small molecule SU11274. Our results reveal a distinct inhibition pattern of the four mutations with IC(50) values for autophosphorylation inhibition ranging between 0.15 and 1.5muM. Differences were further seen on the ability of SU11274 to inhibit phosphorylation of downstream MET transducers such as AKT, ERK, PLCgamma and STAT3 and a variety of MET-dependent biological endpoints. In all the assays, H1112L was the most sensitive to SU11274, while V1206L was less affected under the used concentration range. The differences in responses to SU11274 are discussed based on a structural model of the MET kinase domain.


Asunto(s)
Indoles/farmacología , Mutación/efectos de los fármacos , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-met/química , Proteínas Proto-Oncogénicas c-met/genética , Sulfonamidas/farmacología , Animales , Western Blotting , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ratones , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación/genética , Células 3T3 NIH , Proteína Oncogénica v-akt/metabolismo , Fosfolipasa C gamma/metabolismo , Fosforilación/efectos de los fármacos , Conformación Proteica , Proteínas Proto-Oncogénicas c-met/metabolismo , Factor de Transcripción STAT3/metabolismo , Cicatrización de Heridas/efectos de los fármacos
4.
Genes Cancer ; 1(10): 1053-62, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21779429

RESUMEN

While recent studies implicate that signaling through the receptor tyrosine kinase MET protects cancer cells from DNA damage, molecular events linking MET to the DNA damage response machinery are largely unknown. Here, we studied the impact of MET inhibition by the small molecule PHA665752 on cytotoxicity induced by DNA-damaging agents. We demonstrate that PHA665752 reduces clonogenic survival of tumor cells with MET overexpression when combined with ionizing radiation and synergistically cooperates with ionizing radiation or adriamycin to induce apoptosis. In search of mechanisms underlying the observed synergism, we show that PHA665752 alone considerably increases γH2AX levels, indicating the accumulation of double-strand DNA breaks. In addition, PHA665752 treatment results in sustained high levels of γH2AX and phosphorylated ATM postirradiation, strengthening the assumption that MET inhibition attenuates postdamage DNA repair. PHA665752, alone or in combination with irradiation, leads also to a massive increase of γH2AX tyrosine phosphorylation and its subsequent interaction with the proapoptotic kinase JNK1. Finally, MET inhibition reduces activation of ATR, CHK1, and CDC25B and abrogates an associated DNA damage-induced S phase arrest. This indicates that MET inhibition compromises a critical damage-dependent checkpoint that may enable DNA-damaged cells to exit cell cycle arrest before repair is completed.

5.
Clin Exp Metastasis ; 26(7): 809-15, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19639388

RESUMEN

We investigated if the MET-activating point mutation Y1253D influences clinical outcomes in patients with advanced squamous cell carcinoma of the head and neck (HNSCC). The study population consisted of 152 HNSCC patients treated by hyperfractionated radiotherapy alone or concomitant with chemotherapy between September 1994 and July 2000. Tumors were screened for the presence of the MET-activating point mutation Y1253D. Seventy-eight patients (51%) received radiotherapy alone, 74 patients (49%) underwent radiotherapy concomitant with chemotherapy. Median patient age was 54 years and median follow-up was 5.5 years. Distant metastasis-free survival, local relapse-free survival and overall survival were compared with MET Y1253D status. During follow-up, 29 (19%) patients developed distant metastasis. MET Y1253D was detected in tumors of 21 out of 152 patients (14%). Distant metastasis-free survival (P = 0.008) was associated with MET Y1253D. In a multivariate Cox regression model, adjusted for T-category, only presence of MET Y1253D was associated with decreased distant metastasis-free survival: hazard ratio = 2.5 (95% confidence interval: 1.1, 5.8). The observed association between MET Y1253D-activating point mutation and decreased distant metastasis-free survival in advanced HNSCC suggests that MET may be a potential target for specific treatment interventions.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/patología , Metástasis de la Neoplasia/genética , Mutación Puntual , Proteínas Proto-Oncogénicas/genética , Receptores de Factores de Crecimiento/genética , Secuencia de Bases , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Cartilla de ADN , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-met
6.
Cancer Res ; 68(14): 5769-77, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632630

RESUMEN

Abnormal activation of DNA repair pathways by deregulated signaling of receptor tyrosine kinase systems is a compelling likelihood with significant implications in both cancer biology and treatment. Here, we show that due to a potential substrate switch, mutated variants of the receptor for hepatocyte growth factor Met, but not the wild-type form of the receptor, directly couple to the Abl tyrosine kinase and the Rad51 recombinase, two key signaling elements of homologous recombination-based DNA repair. Treatment of cells that express the mutated receptor variants with the Met inhibitor SU11274 leads, in a mutant-dependent manner, to a reduction of tyrosine phosphorylated levels of Abl and Rad51, impairs radiation-induced nuclear translocation of Rad51, and acts as a radiosensitizer together with the p53 inhibitor pifithrin-alpha by increasing cellular double-strand DNA break levels following exposure to ionizing radiation. Finally, we propose that in order to overcome a mutation-dependent resistance to SU11274, this aberrant molecular axis may alternatively be targeted with the Abl inhibitor, nilotinib.


Asunto(s)
Reparación del ADN , Genes abl , Variación Genética , Mutación , Recombinasa Rad51/genética , Transporte Activo de Núcleo Celular , Animales , Benzotiazoles/metabolismo , Daño del ADN , Indoles/farmacología , Ratones , Células 3T3 NIH , Fosforilación , Piperazinas/farmacología , Recombinasa Rad51/fisiología , Fármacos Sensibilizantes a Radiaciones/farmacología , Sulfonamidas/farmacología , Tolueno/análogos & derivados , Tolueno/metabolismo , Tirosina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA