Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Clin Exp Med ; 19(2): 201-210, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30523507

RESUMEN

The purpose of the present study was to investigate the in vitro and in vivo activity of PLX9486, a tyrosine kinase inhibitor (TKI) targeting both primary KIT exon 9 and 11 and secondary exon 17 and 18 mutations in gastrointestinal stromal tumors (GISTs). Imatinib, a potent inhibitor of mutated KIT, has revolutionized the clinical management of advanced, metastatic GIST. However, secondary resistance develops mainly through acquired mutations in KIT exons 13/14 or exons 17/18. Second-line sunitinib potently inhibits KIT exon 13/14 mutants but is ineffective against exon 17 mutations. In our study, PLX9486 demonstrated in vitro nanomolar potency in inhibiting the growth and KIT phosphorylation of engineered BaF3 cells transformed with KIT exon 17 mutations (p.D816V) and with the double KIT exon 11/17 mutations (p.V560G/D816V). The in vivo efficacy of PLX9486 was evaluated using two imatinib-resistant GIST patient-derived xenograft (PDX) models. In UZLX-GIST9 (KIT: p.P577del;W557LfsX5;D820G), PLX9486 100 mg/kg/day resulted in significant inhibition of proliferation. Pharmacodynamic analysis showed a pronounced reduction in mitogen-activated protein kinase (MAPK) activation and other downstream effects of the KIT signaling pathway but no significant effect on KIT Y703 and Y719 phosphorylation. Similarly, in MRL-GIST1 (KIT: p.W557_K558del;Y823D) PLX9486 treatment led to significant tumor regression and strong inhibition of MAPK activation. Interestingly, the inhibitory effect on MAPK activation was evident even after a single dose of PLX9486. In conclusion, PLX9486 showed anti-tumor efficacy in patient-derived imatinib-resistant GIST xenograft models, mainly through inhibition of KIT signaling. These preclinical efficacy data encourage further testing of PLX9486 in the clinical setting.


Asunto(s)
Antineoplásicos/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/patología , Proteínas Mutantes/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Xenoinjertos , Humanos , Ratones , Proteínas Mutantes/metabolismo , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-kit/metabolismo , Resultado del Tratamiento
2.
Pigment Cell Melanoma Res ; 26(6): 845-51, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23890088

RESUMEN

BRAF is the most prevalent oncogene and an important therapeutic target in melanoma. In some cancers, BRAF is activated by rearrangements that fuse its kinase domain to 5' partner genes. We examined 848 comparative genomic hybridization profiles of melanocytic tumors and found copy number transitions within BRAF in 10 tumors, of which six could be further characterized by sequencing. In all, the BRAF kinase domain was fused in-frame to six N-terminal partners. No other mutations were identified in melanoma oncogenes. One of the seven melanoma cell lines without known oncogenic mutations harbored a similar BRAF fusion, which constitutively activated the MAP kinase pathway. Sorafenib, but not vemurafenib, could block MAP kinase pathway activation and proliferation of the cell line at clinically relevant concentrations, whereas BRAF(V) (600E) mutant melanoma cell lines were significantly more sensitive to vemurafenib. The patient from whom the cell line was derived showed a durable clinical response to sorafenib.


Asunto(s)
Melanocitos/patología , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Terapia Molecular Dirigida , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/enzimología , Adolescente , Adulto , Preescolar , Activación Enzimática/efectos de los fármacos , Femenino , Reordenamiento Génico/efectos de los fármacos , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Melanocitos/efectos de los fármacos , Melanocitos/enzimología , Melanoma/patología , Persona de Mediana Edad , Nevo de Células Epitelioides y Fusiformes/patología , Niacinamida/análogos & derivados , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Sorafenib , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Vemurafenib , Adulto Joven
3.
Curr Top Microbiol Immunol ; 355: 3-18, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21809194

RESUMEN

Discovery of novel kinase inhibitors has matured rapidly over the last decade. Paramount to the successful development of kinase inhibitors is appropriate selectivity for validated targets. Many different approaches have been applied over the years, with varied results. There are currently thirteen different small molecule protein kinase inhibitors on the marketplace. Interestingly, a majority of these compounds lack precise selectivity for specific targets. This will change in the coming years, as technology for achieving improved selectivity becomes more widely applied. This chapter will focus on some of the critical considerations in setting up a kinase discovery and development project, citing examples particularly targeting the Raf kinases.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores de Proteínas Quinasas/farmacología , Quinasas raf/antagonistas & inhibidores , Adenosina Trifosfato/química , Animales , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Activación Enzimática , Pruebas de Enzimas/métodos , Humanos , Indoles/farmacología , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Ratones , Niacinamida/análogos & derivados , Niacinamida/química , Niacinamida/farmacología , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Fosforilación , Inhibidores de Proteínas Quinasas/química , Solubilidad , Sorafenib , Relación Estructura-Actividad , Sulfonamidas/farmacología , Quinasas raf/química
4.
Neoplasia ; 12(8): 637-49, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20689758

RESUMEN

PURPOSE: About 65% to 70% of melanomas harbor a mutation in v-raf murine sarcoma viral oncogene homolog B1 (BRAF) that causes the steady-state activation of extracellular signal-regulated kinase (ERK). We sought to investigate the efficacy of PLX4032 (BRAF inhibitor) to identify patterns/predictors of response/resistance and to study the effects of BRAF in melanoma. EXPERIMENTAL DESIGN: Well-characterized melanoma cell lines, including several with acquired drug resistance, were exposed to PLX4032. Growth inhibition, phosphosignaling, cell cycle, apoptosis, and gene expression analyses were performed before and after exposure to drug. RESULTS: Using a growth-adjusted inhibitory concentration of 50% cutoff of 1 microM, 13 of 35 cell lines were sensitive to PLX4032, 16 resistant, and 6 intermediate (37%, 46%, and 17% respectively). PLX4032 caused growth inhibition, G(0)/G(1) arrest, and restored apoptosis in the sensitive cell lines. A BRAF mutation predicted for but did not guarantee a response, whereas a neuroblastoma RAS viral oncogene homolog mutation or wild-type BRAF conferred resistance. Cells with concurrent BRAF mutations and melanocortin 1 receptor germ line variants and/or a more differentiated melanocyte genotype had a preferential response. Acquired PLX4032 resistance reestablishes ERK signaling, promotes a nonmelanocytic genotype, and is associated with an increase in the gene expression of certain metallothioneins and mediators of angiogenesis. CONCLUSIONS: PLX4032 has robust activity in BRAF mutated melanoma. The preclinical use of this molecule identifies criteria for its proper clinical application, describes patterns of and reasons for response/resistance, and affords insight into the role of a BRAF mutation in melanoma.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Indoles/farmacocinética , Melanoma/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/metabolismo , Sulfonamidas/farmacocinética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biomarcadores Farmacológicos/análisis , Biomarcadores Farmacológicos/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Análisis Mutacional de ADN , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Melanoma/genética , Melanoma/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Especificidad por Sustrato/efectos de los fármacos , Sulfonamidas/farmacología , Resultado del Tratamiento , Vemurafenib
5.
J Clin Endocrinol Metab ; 95(1): 450-5, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19880792

RESUMEN

CONTEXT: The V600E mutation accounts for the vast majority of thyroid carcinoma-associated BRAF mutations. OBJECTIVE: The aim was to study the effects of the two BRAF V600E ATP-competitive kinase inhibitors, PLX4032 and PLX4720, in thyroid carcinoma cell lines. EXPERIMENTAL DESIGN: We examined the activity of PLX4032 and PLX4720 in thyroid carcinoma cell lines harboring BRAF V600E (8505C, BCPAP, SW1736, BHT101), NRAS Q61R (HTH7), KRAS G12R (CAL62), HRAS G13R (C643), or RET/PTC1 (TPC-1) oncogenes. Normal thyrocytes (PC Cl 3) were used as control. RESULTS: Both compounds inhibited the proliferation of BRAF mutant cell lines, but not normal thyrocytes, with a half maximal effective concentration (EC(50)) ranging from 78-113 nm for PLX4720 and from 29-97 nm for PLX4032. Doses equal to or higher than 500 nm were required to achieve a similar effect in BRAF wild-type cancer cells. Phosphorylation of ERK 1/2 and MAPK kinase (MEK) 1/2 decreased upon PLX4032 and PLX4720 treatment in BRAF mutant thyroid carcinoma cells but not in normal thyroid cells or in cell lines harboring mutations of RAS or RET/PTC1 rearrangements. PLX4032 and PLX4720 treatment induced a G(1) block and altered expression of genes involved in the control of G(1)-S cell-cycle transition. 8505C cell tumor xenografts were smaller in nude mice treated with PLX4032 than in control mice. This inhibition was associated with reduction of phospho-ERK and phospho-MEK levels. CONCLUSIONS: This study provides additional evidence of the promising nature of mutant BRAF as a molecular target for thyroid carcinoma cells.


Asunto(s)
Carcinoma/patología , Citostáticos/farmacología , Indoles/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/farmacología , Neoplasias de la Tiroides/patología , Adenosina Trifosfato/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Unión Competitiva/efectos de los fármacos , Carcinoma/genética , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Suero/fisiología , Neoplasias de la Tiroides/genética , Células Tumorales Cultivadas
6.
Clin Cancer Res ; 15(16): 5032-5039, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19671848

RESUMEN

PURPOSE: Neurofibromatosis type 2 (NF2) is a rare autosomal dominant disorder associated primarily with bilateral schwannomas seen on the superior vestibular branches of the eighth cranial nerves. Significant morbidity can result from surgical treatment of these tumors. Meningiomas, ependymomas, and other benign central nervous system tumors are also common in NF2. The lack of effective treatments for NF2 marks an unmet medical need. EXPERIMENTAL DESIGN: Here, we provide recommendations from a workshop, cochaired by Drs. D. Gareth Evans and Marco Giovannini, of 36 international researchers, physicians, representatives of the biotechnology industry, and patient advocates on how to accelerate progress toward NF2 clinical trials. RESULTS: Workshop participants reached a consensus that, based on current knowledge, the time is right to plan and implement NF2 clinical trials. Obstacles impeding NF2 clinical trials and how to address them were discussed, as well as the candidate therapeutic pipeline for NF2. CONCLUSIONS: Both phase 0 and phase II NF2 trials are near-term options for NF2 clinical trials. The number of NF2 patients in the population remains limited, and successful recruitment will require ongoing collaboration efforts between NF2 clinics.


Asunto(s)
Ensayos Clínicos como Asunto/métodos , Consenso , Directrices para la Planificación en Salud , Neurofibromatosis 2/terapia , Animales , Implantes Auditivos de Tronco Encefálico , Implantes Cocleares , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/tendencias , Humanos , Neoplasias Meníngeas/terapia , Meningioma/terapia , Neurofibromatosis 2/diagnóstico , Factores de Tiempo
7.
Curr Opin Investig Drugs ; 4(12): 1436-41, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14763129

RESUMEN

Recognition of the importance of the Raf pathway in the proliferation and survival of tumor cells recently increased with the discovery of activating BRAF mutations in human tumors. Therefore, in addition to a role in controlling tumors with Ras mutations and activated growth factor receptors, inhibitors of the Raf pathway may harbor therapeutic potential in tumors carrying a BRAF oncogene. A variety of agents have been discovered that interfere with the Raf pathway, including antisense oligonucleotides and small molecules. These inhibitors block the expression of Raf protein, block Ras/Raf interaction, block its kinase activity, or block the kinase activity of the Raf target protein mitogen-activated protein kinase kinase. Raf pathway inhibitors that are currently undergoing clinical evaluation show promising signs of anticancer efficacy with a very tolerable safety profile. Indeed, the Raf inhibitor BAY-43-9006 recently entered phase III clinical trials. Here, we review the current development status of potential Raf pathway therapeutics.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Bencenosulfonatos/química , Bencenosulfonatos/farmacología , Bencenosulfonatos/uso terapéutico , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias/enzimología , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Piridinas/química , Piridinas/farmacología , Piridinas/uso terapéutico , Sorafenib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA