Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
ACS Med Chem Lett ; 12(1): 93-98, 2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33488969

RESUMEN

Fibroblast growth factor receptors (FGFR) 2 and 3 have been established as drivers of numerous types of cancer with multiple drugs approved or entering late stage clinical trials. A limitation of current inhibitors is vulnerability to gatekeeper resistance mutations. Using a combination of targeted high-throughput screening and structure-based drug design, we have developed a series of aminopyrazole based FGFR inhibitors that covalently target a cysteine residue on the P-loop of the kinase. The inhibitors show excellent activity against the wild-type and gatekeeper mutant versions of the enzymes. Further optimization using SAR analysis and structure-based drug design led to analogues with improved potency and drug metabolism and pharmacokinetics properties.

2.
Mol Cancer Ther ; 7(9): 2955-66, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18790776

RESUMEN

Insights from cell cycle research have led to the hypothesis that tumors may be selectively sensitized to DNA-damaging agents resulting in improved antitumor activity and a wider therapeutic margin. The theory relies on the observation that the majority of tumors are deficient in the G1-DNA damage checkpoint pathway resulting in reliance on S and G2 checkpoints for DNA repair and cell survival. The S and G2 checkpoints are regulated by checkpoint kinase 1, a serine/threonine kinase that is activated in response to DNA damage; thus, inhibition of checkpoint kinase 1 signaling impairs DNA repair and increases tumor cell death. Normal tissues, however, have a functioning G1 checkpoint signaling pathway allowing for DNA repair and cell survival. Here, we describe the preclinical profile of AZD7762, a potent ATP-competitive checkpoint kinase inhibitor in clinical trials. AZD7762 has been profiled extensively in vitro and in vivo in combination with DNA-damaging agents and has been shown to potentiate response in several different settings where inhibition of checkpoint kinase results in the abrogation of DNA damage-induced cell cycle arrest. Dose-dependent potentiation of antitumor activity, when AZD7762 is administered in combination with DNA-damaging agents, has been observed in multiple xenograft models with several DNA-damaging agents, further supporting the potential of checkpoint kinase inhibitors to enhance the efficacy of both conventional chemotherapy and radiotherapy and increase patient response rates in a variety of settings.


Asunto(s)
Daño del ADN , ADN de Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Tiofenos/farmacología , Urea/análogos & derivados , Animales , Bioensayo , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sinergismo Farmacológico , Fase G2/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos , Masculino , Ratones , Mutación/genética , Inhibidores de Proteínas Quinasas/análisis , Inhibidores de Proteínas Quinasas/química , Ratas , Tiofenos/análisis , Tiofenos/química , Topotecan/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Urea/análisis , Urea/química , Urea/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
3.
Nat Med ; 24(4): 497-504, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29457796

RESUMEN

Genomic analyses of cancer have identified recurrent point mutations in the RNA splicing factor-encoding genes SF3B1, U2AF1, and SRSF2 that confer an alteration of function. Cancer cells bearing these mutations are preferentially dependent on wild-type (WT) spliceosome function, but clinically relevant means to therapeutically target the spliceosome do not currently exist. Here we describe an orally available modulator of the SF3b complex, H3B-8800, which potently and preferentially kills spliceosome-mutant epithelial and hematologic tumor cells. These killing effects of H3B-8800 are due to its direct interaction with the SF3b complex, as evidenced by loss of H3B-8800 activity in drug-resistant cells bearing mutations in genes encoding SF3b components. Although H3B-8800 modulates WT and mutant spliceosome activity, the preferential killing of spliceosome-mutant cells is due to retention of short, GC-rich introns, which are enriched for genes encoding spliceosome components. These data demonstrate the therapeutic potential of splicing modulation in spliceosome-mutant cancers.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/genética , Piperazinas/farmacología , Piridinas/farmacología , Empalme del ARN/genética , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Empalmosomas/genética , Administración Oral , Animales , Secuencia de Bases , Humanos , Intrones/genética , Células K562 , Leucemia/genética , Leucemia/patología , Ratones , Mutación , Neoplasias/patología , Piperazinas/administración & dosificación , Piridinas/administración & dosificación , Empalme del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Discov ; 8(9): 1176-1193, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29991605

RESUMEN

Mutations in estrogen receptor alpha (ERα) that confer resistance to existing classes of endocrine therapies are detected in up to 30% of patients who have relapsed during endocrine treatments. Because a significant proportion of therapy-resistant breast cancer metastases continue to be dependent on ERα signaling, there remains a critical need to develop the next generation of ERα antagonists that can overcome aberrant ERα activity. Through our drug-discovery efforts, we identified H3B-5942, which covalently inactivates both wild-type and mutant ERα by targeting Cys530 and enforcing a unique antagonist conformation. H3B-5942 belongs to a class of ERα antagonists referred to as selective estrogen receptor covalent antagonists (SERCA). In vitro comparisons of H3B-5942 with standard-of-care (SoC) and experimental agents confirmed increased antagonist activity across a panel of ERαWT and ERαMUT cell lines. In vivo, H3B-5942 demonstrated significant single-agent antitumor activity in xenograft models representing ERαWT and ERαY537S breast cancer that was superior to fulvestrant. Lastly, H3B-5942 potency can be further improved in combination with CDK4/6 or mTOR inhibitors in both ERαWT and ERαMUT cell lines and/or tumor models. In summary, H3B-5942 belongs to a class of orally available ERα covalent antagonists with an improved profile over SoCs.Significance: Nearly 30% of endocrine therapy-resistant breast cancer metastases harbor constitutively activating mutations in ERα. SERCA H3B-5942 engages C530 of both ERαWT and ERαMUT, promotes a unique antagonist conformation, and demonstrates improved in vitro and in vivo activity over SoC agents. Importantly, single-agent efficacy can be further enhanced by combining with CDK4/6 or mTOR inhibitors. Cancer Discov; 8(9); 1176-93. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 1047.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Antagonistas del Receptor de Estrógeno/administración & dosificación , Receptor alfa de Estrógeno/antagonistas & inhibidores , Indazoles/administración & dosificación , Mutación , Administración Oral , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisteína/antagonistas & inhibidores , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Antagonistas del Receptor de Estrógeno/química , Antagonistas del Receptor de Estrógeno/farmacología , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Indazoles/química , Indazoles/farmacología , Células MCF-7 , Ratones , Conformación Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Commun ; 8: 15522, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28541300

RESUMEN

Pladienolide, herboxidiene and spliceostatin have been identified as splicing modulators that target SF3B1 in the SF3b subcomplex. Here we report that PHF5A, another component of this subcomplex, is also targeted by these compounds. Mutations in PHF5A-Y36, SF3B1-K1071, SF3B1-R1074 and SF3B1-V1078 confer resistance to these modulators, suggesting a common interaction site. RNA-seq analysis reveals that PHF5A-Y36C has minimal effect on basal splicing but inhibits the global action of splicing modulators. Moreover, PHF5A-Y36C alters splicing modulator-induced intron-retention/exon-skipping profile, which correlates with the differential GC content between adjacent introns and exons. We determine the crystal structure of human PHF5A demonstrating that Y36 is located on a highly conserved surface. Analysis of the cryo-EM spliceosome Bact complex shows that the resistance mutations cluster in a pocket surrounding the branch point adenosine, suggesting a competitive mode of action. Collectively, we propose that PHF5A-SF3B1 forms a central node for binding to these splicing modulators.


Asunto(s)
Adenosina/química , Empalme Alternativo , Proteínas Portadoras/química , Fosfoproteínas/química , Factores de Empalme de ARN/química , Proliferación Celular , Supervivencia Celular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Compuestos Epoxi/química , Exones , Alcoholes Grasos/química , Células HCT116 , Humanos , Intrones , Macrólidos/química , Espectrometría de Masas , Mutagénesis Sitio-Dirigida , Mutación , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Fosfoproteínas/metabolismo , Unión Proteica , Conformación Proteica , Piranos/química , Interferencia de ARN , Factores de Empalme de ARN/metabolismo , Proteínas de Unión al ARN , Proteínas Recombinantes/química , Análisis de Secuencia de ARN , Compuestos de Espiro/química , Empalmosomas/metabolismo , Transactivadores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA