Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
N Engl J Med ; 366(3): 207-15, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22256804

RESUMEN

BACKGROUND: Cutaneous squamous-cell carcinomas and keratoacanthomas are common findings in patients treated with BRAF inhibitors. METHODS: We performed a molecular analysis to identify oncogenic mutations (HRAS, KRAS, NRAS, CDKN2A, and TP53) in the lesions from patients treated with the BRAF inhibitor vemurafenib. An analysis of an independent validation set and functional studies with BRAF inhibitors in the presence of the prevalent RAS mutation was also performed. RESULTS: Among 21 tumor samples, 13 had RAS mutations (12 in HRAS). In a validation set of 14 samples, 8 had RAS mutations (4 in HRAS). Thus, 60% (21 of 35) of the specimens harbored RAS mutations, the most prevalent being HRAS Q61L. Increased proliferation of HRAS Q61L-mutant cell lines exposed to vemurafenib was associated with mitogen-activated protein kinase (MAPK)-pathway signaling and activation of ERK-mediated transcription. In a mouse model of HRAS Q61L-mediated skin carcinogenesis, the vemurafenib analogue PLX4720 was not an initiator or a promoter of carcinogenesis but accelerated growth of the lesions harboring HRAS mutations, and this growth was blocked by concomitant treatment with a MEK inhibitor. CONCLUSIONS: Mutations in RAS, particularly HRAS, are frequent in cutaneous squamous-cell carcinomas and keratoacanthomas that develop in patients treated with vemurafenib. The molecular mechanism is consistent with the paradoxical activation of MAPK signaling and leads to accelerated growth of these lesions. (Funded by Hoffmann-La Roche and others; ClinicalTrials.gov numbers, NCT00405587, NCT00949702, NCT01001299, and NCT01006980.).


Asunto(s)
Carcinoma de Células Escamosas/genética , Genes ras , Indoles/uso terapéutico , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/genética , Sulfonamidas/uso terapéutico , Anciano , Anciano de 80 o más Años , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Indoles/administración & dosificación , Masculino , Ratones , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Sulfonamidas/administración & dosificación , Vemurafenib
2.
Sci Rep ; 14(1): 2647, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38302613

RESUMEN

This study aims to evaluate the effect of social capital (SC), social support (SS), and social network formation (SNF) on the quality of life of American adults during COVID-19. Using a probability sample of American adults aged 49+, 2370 respondents were selected from the National Social Life Health and Aging Project (NSHAP) dataset for analysis using an integrated partial least squares based on structural equation modeling (PLS-SEM)-K-fold cross-validation approach. The analysis showed that social capital assessed using civic engagement, social cohesion, socioeconomic status (SES), social support, and social network formation were significantly and positively associated with American adults' quality of life during the COVID-19 pandemic. Furthermore, the results showed that using the PLS-SEM and K-fold cross-validation approach produced a medium predictive power of the overall model, confirming the importance of SC, SS, and SNF in predicting quality of life-outcomes. These findings suggest that efforts to promote the well-being of American adults, especially older adults, during the pandemic should focus on strengthening social capital, social support and social network formation.


Asunto(s)
COVID-19 , Capital Social , Humanos , Estados Unidos/epidemiología , Anciano , Calidad de Vida , Pandemias , COVID-19/epidemiología , Apoyo Social , Clase Social , Red Social
3.
Angiogenesis ; 15(4): 623-41, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22843200

RESUMEN

Sunitinib is a potent and clinically approved tyrosine kinase inhibitor that can suppress tumour growth by inhibiting angiogenesis. However, conflicting data exist regarding the effects of this drug on the growth of metastases in preclinical models. Here we use 4T1 and RENCA tumour cells, which both form lung metastases in Balb/c mice, to re-address the effects of sunitinib on the progression of metastatic disease in mice. We show that treatment of mice with sunitinib prior to intravenous injection of tumour cells can promote the seeding and growth of 4T1 lung metastases, but not RENCA lung metastases, showing that this effect is cell line dependent. However, increased metastasis occurred only upon administration of a very high sunitinib dose, but not when lower, clinically relevant doses were used. Mechanistically, high dose sunitinib led to a pericyte depletion effect in the lung vasculature that correlated with increased seeding of metastasis. By administering sunitinib to mice after intravenous injection of tumour cells, we demonstrate that while sunitinib does not inhibit the growth of 4T1 lung tumour nodules, it does block the growth of RENCA lung tumour nodules. This contrasting response was correlated with increased myeloid cell recruitment and persistent vascularisation in 4T1 tumours, whereas RENCA tumours recruited less myeloid cells and were more profoundly devascularised upon sunitinib treatment. Finally, we show that progression of 4T1 tumours in sunitinib treated mice results in increased hypoxia and increased glucose metabolism in these tumours and that this is associated with a poor outcome. Taken together, these data suggest that the effects of sunitinib on tumour progression are dose-dependent and tumour model-dependent. These findings have relevance for understanding how anti-angiogenic agents may influence disease progression when used in the adjuvant or metastatic setting in cancer patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Indoles/uso terapéutico , Metástasis de la Neoplasia/tratamiento farmacológico , Pirroles/uso terapéutico , Animales , Ratones , Ratones Endogámicos BALB C , Tomografía de Emisión de Positrones , Sunitinib , Tomografía Computarizada por Rayos X
4.
Mol Cancer Ther ; 15(1): 172-83, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26487278

RESUMEN

Sunitinib and pazopanib are antiangiogenic tyrosine kinase inhibitors (TKI) used to treat metastatic renal cell carcinoma (RCC). However, the ability of these drugs to extend progression-free and overall survival in this patient population is limited by drug resistance. It is possible that treatment outcomes in RCC patients could be improved by rationally combining TKIs with other agents. Here, we address whether inhibition of the Ras-Raf-MEK-ERK1/2 pathway is a rational means to improve the response to TKIs in RCC. Using a xenograft model of RCC, we found that tumors that are resistant to sunitinib have a significantly increased angiogenic response compared with tumors that are sensitive to sunitinib in vivo. We also observed significantly increased levels of phosphorylated ERK1/2 in the vasculature of resistant tumors, when compared with sensitive tumors. These data suggested that the Ras-Raf-MEK-ERK1/2 pathway, an important driver of angiogenesis in endothelial cells, remains active in the vasculature of TKI-resistant tumors. Using an in vitro angiogenesis assay, we identified that the MEK inhibitor (MEKI) trametinib has potent antiangiogenic activity. We then show that, when trametinib is combined with a TKI in vivo, more effective suppression of tumor growth and tumor angiogenesis is achieved than when either drug is utilized alone. In conclusion, we provide preclinical evidence that combining a TKI, such as sunitinib or pazopanib, with a MEKI, such as trametinib, is a rational and efficacious treatment regimen for RCC.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Inhibidores de Proteínas Quinasas/farmacología , Piridonas/farmacología , Pirimidinonas/farmacología , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Indoles/farmacología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Pirroles/farmacología , Sunitinib , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Cell ; 27(1): 85-96, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25500121

RESUMEN

BRAF and MEK inhibitors are effective in BRAF mutant melanoma, but most patients eventually relapse with acquired resistance, and others present intrinsic resistance to these drugs. Resistance is often mediated by pathway reactivation through receptor tyrosine kinase (RTK)/SRC-family kinase (SFK) signaling or mutant NRAS, which drive paradoxical reactivation of the pathway. We describe pan-RAF inhibitors (CCT196969, CCT241161) that also inhibit SFKs. These compounds do not drive paradoxical pathway activation and inhibit MEK/ERK in BRAF and NRAS mutant melanoma. They inhibit melanoma cells and patient-derived xenografts that are resistant to BRAF and BRAF/MEK inhibitors. Thus, paradox-breaking pan-RAF inhibitors that also inhibit SFKs could provide first-line treatment for BRAF and NRAS mutant melanomas and second-line treatment for patients who develop resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Pirazinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanoma/patología , Melanoma Experimental , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 70(20): 8036-44, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20807807

RESUMEN

Oncogenic BRAF is a critical driver of proliferation and survival and is thus a validated therapeutic target in cancer. We have developed a potent inhibitor, termed 1t (CCT239065), of the mutant protein kinase, (V600E)BRAF. 1t inhibits signaling downstream of (V600E)BRAF in cancer cells, blocking DNA synthesis, and inhibiting proliferation. Importantly, we show that 1t is considerably more selective for mutated BRAF cancer cell lines compared with wild-type BRAF lines. The inhibitor is well tolerated in mice and exhibits excellent oral bioavailability (F = 71%). Suppression of (V600E)BRAF-mediated signaling in human tumor xenografts was observed following oral administration of a single dose of 1t. As expected, the growth rate in vivo of a wild-type BRAF human tumor xenograft model is unaffected by inhibitor 1t. In contrast, 1t elicits significant therapeutic responses in mutant BRAF-driven human melanoma xenografts.


Asunto(s)
Melanoma/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas B-raf/genética , Administración Oral , Sustitución de Aminoácidos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , División Celular , Línea Celular Tumoral , Supervivencia Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Hibridación de Ácido Nucleico , Fosforilación , Trasplante Heterólogo
8.
J Med Chem ; 53(15): 5639-55, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20597484

RESUMEN

Mutated BRAF serine/threonine kinase is implicated in several types of cancer, with particularly high frequency in melanoma and colorectal carcinoma. We recently reported on the development of BRAF inhibitors based on a tripartite A-B-C system featuring an imidazo[4,5]pyridin-2-one group hinge binder. Here we present the design, synthesis, and optimization of a new series of inhibitors with a different A-B-C system that has been modified by the introduction of a range of novel hinge binders (A ring). The optimization of the hinge binding moiety has enabled the development of compounds with low nanomolar potencies in both BRAF inhibition and cellular assays. These compounds display optimal pharmacokinetic properties that warrant further in vivo investigations.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Pirazinas/síntesis química , Piridinas/síntesis química , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Bencenosulfonatos/química , Disponibilidad Biológica , Cristalografía por Rayos X , Femenino , Humanos , Enlace de Hidrógeno , Imidazoles/química , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Trasplante de Neoplasias , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Unión Proteica , Proteínas Proto-Oncogénicas B-raf/química , Pirazinas/farmacocinética , Pirazinas/farmacología , Piridinas/química , Piridinas/farmacocinética , Piridinas/farmacología , Sorafenib , Relación Estructura-Actividad , Trasplante Heterólogo
9.
J Med Chem ; 53(7): 2741-56, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20199087

RESUMEN

We describe the design, synthesis, and optimization of a series of new inhibitors of V-RAF murine sarcoma viral oncogene homologue B1 (BRAF), a kinase whose mutant form (V600E) is implicated in several types of cancer, with a particularly high frequency in melanoma. Our previously described inhibitors with a tripartite A-B-C system (where A is a hinge binding pyrido[4,5-b]imidazolone system, B is an aryl spacer group, and C is a heteroaromatic group) were potent against purified (V600E)BRAF in vitro but were less potent in accompanying cellular assays. Substitution of different aromatic heterocycles for the phenyl based C-ring is evaluated herein as a potential means of improving the cellular potencies of these inhibitors. Substituted pyrazoles, particularly 3-tert-butyl-1-aryl-1H-pyrazoles, increase the cellular potencies without detrimental effects on the potency on isolated (V600E)BRAF. Thus, compounds have been synthesized that inhibit, with low nanomolar concentrations, (V600E)BRAF, its downstream signaling in cells [as measured by the reduction of the phosphorylation of extracellular regulated kinase (ERK)], and the proliferation of mutant BRAF-dependent cells. Concomitant benefits are good oral bioavailability and high plasma concentrations in vivo.


Asunto(s)
Diseño de Fármacos , Proteínas Oncogénicas v-raf/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Virus del Sarcoma Murino/enzimología , Homología de Secuencia , Animales , Línea Celular Tumoral , Femenino , Humanos , Concentración 50 Inhibidora , Ratones , Modelos Moleculares , Conformación Molecular , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas B-raf/química , Proteínas Proto-Oncogénicas B-raf/metabolismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA