Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(8): 2724-9, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22003129

RESUMEN

Breast cancers are comprised of molecularly distinct subtypes that may respond differently to pathway-targeted therapies now under development. Collections of breast cancer cell lines mirror many of the molecular subtypes and pathways found in tumors, suggesting that treatment of cell lines with candidate therapeutic compounds can guide identification of associations between molecular subtypes, pathways, and drug response. In a test of 77 therapeutic compounds, nearly all drugs showed differential responses across these cell lines, and approximately one third showed subtype-, pathway-, and/or genomic aberration-specific responses. These observations suggest mechanisms of response and resistance and may inform efforts to develop molecular assays that predict clinical response.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Dosificación de Gen/genética , Humanos , Modelos Biológicos , Transducción de Señal/genética , Transcripción Genética/efectos de los fármacos
2.
Proc Natl Acad Sci U S A ; 107(13): 5839-44, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20167803

RESUMEN

Centromere-associated protein-E (CENP-E) is a kinetochore-associated mitotic kinesin that is thought to function as the key receptor responsible for mitotic checkpoint signal transduction after interaction with spindle microtubules. We have identified GSK923295, an allosteric inhibitor of CENP-E kinesin motor ATPase activity, and mapped the inhibitor binding site to a region similar to that bound by loop-5 inhibitors of the kinesin KSP/Eg5. Unlike these KSP inhibitors, which block release of ADP and destabilize motor-microtubule interaction, GSK923295 inhibited release of inorganic phosphate and stabilized CENP-E motor domain interaction with microtubules. Inhibition of CENP-E motor activity in cultured cells and tumor xenografts caused failure of metaphase chromosome alignment and induced mitotic arrest, indicating that tight binding of CENP-E to microtubules is insufficient to satisfy the mitotic checkpoint. Consistent with genetic studies in mice suggesting that decreased CENP-E function can have a tumor-suppressive effect, inhibition of CENP-E induced tumor cell apoptosis and tumor regression.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Sarcosina/análogos & derivados , Sitio Alostérico , Animales , Antineoplásicos/química , Sitios de Unión , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Perros , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Técnicas In Vitro , Cinesinas/antagonistas & inhibidores , Cinesinas/química , Cinesinas/metabolismo , Ratones , Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , Modelos Moleculares , Estructura Molecular , Sarcosina/química , Sarcosina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Target Oncol ; 18(2): 269-285, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36826464

RESUMEN

BACKGROUND: Patients with triple-negative breast cancer (TNBC) expressing the androgen receptor (AR) respond poorly to neoadjuvant chemotherapy, although AR antagonists have shown promising clinical activity, suggesting these tumors are AR-dependent. cAMP responsive element binding protein (CREB)-binding protein (CBP) and p300 are transcriptional co-activators for the AR, a key driver of AR+ breast and prostate cancer, and may provide a novel therapeutic target in AR+ TNBC. OBJECTIVES: The aim of this study was to determine the therapeutic potential of FT-6876, a new CBP/p300 bromodomain inhibitor, in breast cancer models with a range of AR levels in vitro and in vivo. METHODS: Effects of FT-6876 on the CBP/p300 pathway were determined by combining chromatin immunoprecipitation (ChIP) with precision run-on sequencing (PRO-seq) complemented with H3K27 acetylation (Ac) and transcriptional profiling. The antiproliferative effect of FT-6876 was also measured in vitro and in vivo. RESULTS: We describe the discovery of FT-6876, a potent and selective CBP/p300 bromodomain inhibitor. The combination of ChIP and PRO-seq confirmed the reduction in H3K27Ac at specific promoter sites concurrent with a decrease in CBP/p300 on the chromatin and a reduction in nascent RNA and enhancer RNA. This was associated with a time- and concentration-dependent reduction in H3K37Ac associated with a decrease in AR and estrogen receptor (ER) target gene expression. This led to a time-dependent growth inhibition in AR+ models, correlated with AR expression. Tumor growth inhibition was also observed in AR+ tumor models of TNBC and ER+ breast cancer subtypes with consistent pharmacokinetics and pharmacodynamics. CONCLUSION: Our findings demonstrate FT-6876 as a promising new CBP/p300 bromodomain inhibitor, with efficacy in preclinical models of AR+ breast cancer.


Asunto(s)
Receptores Androgénicos , Neoplasias de la Mama Triple Negativas , Masculino , Humanos , Receptores Androgénicos/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Unión Proteica , ARN/metabolismo
5.
Biochemistry ; 48(40): 9503-15, 2009 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-19719327

RESUMEN

Structural changes in the mitotic arrest deficient protein 2 (Mad2) have been proposed to be essential for spindle checkpoint function. Current models for checkpoint activation propose that a C-Mad2-Mad1 core complex at unattached kinetochores is required for the structural activation through a process involving the interaction of two Mad2 conformers: a closed conformer bound to Mad1 or Cdc20 and an open conformer unbound to these ligands. To gain a molecular understanding of the mechanisms that accelerate the structural transition between the open and closed Mad2 conformations, we constructed a unique in vitro homogeneous Mad2 activity assay that specifically reports C-Mad2-Cdc20 formation. Using this assay we were are able to directly establish that (a) O-Mad2 transforms into a C-Mad2-Cdc20 complex >300-fold slower than unliganded C-Mad2, (b) a stable C-Mad2-Mad1 core complex catalyzes the transformation of O-Mad2 into a Cdc20-bound C-Mad2 complex, (c) a C-Mad2-Cdc20 complex can promote its own transformation of O-Mad2 into a Cdc20-bound C-Mad2 complex, and (d) the binding interaction between unliganded C-Mad2 and Cdc20 cannot be catalyzed by a C-Mad2-Mad1 core complex. Our data are consistent with the "Mad2 template" catalytic model in which a C-Mad2 template facilitates the binding of O-Mad2 to Cdc20 and supports a mechanism of C-Mad2-Cdc20 formation away from Mad1 containing kinetochores. Furthermore, our unique homogeneous Mad2 assay could be translated into a screening platform to identify small molecule drug-like compounds that directly modulate C-Mad2-Cdc20 formation.


Asunto(s)
Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Huso Acromático/química , Huso Acromático/metabolismo , Secuencia de Aminoácidos , Catálisis , Proteínas Cdc20 , Polarización de Fluorescencia , Humanos , Cinética , Ligandos , Proteínas Mad2 , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Conformación Proteica
6.
Nat Chem Biol ; 3(11): 722-6, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17922005

RESUMEN

The mitotic kinesin KSP (kinesin spindle protein, or Eg5) has an essential role in centrosome separation and formation of the bipolar mitotic spindle. Its exclusive involvement in the mitotic spindle of proliferating cells presents an opportunity for developing new anticancer agents with reduced side effects relative to antimitotics that target tubulin. Ispinesib is an allosteric small-molecule KSP inhibitor in phase 2 clinical trials. Mutations that attenuate ispinesib binding to KSP have been identified, which highlights the need for inhibitors that target different binding sites. We describe a new class of selective KSP inhibitors that are active against ispinesib-resistant forms of KSP. These ATP-competitive KSP inhibitors do not bind in the nucleotide binding pocket. Cumulative data from generation of resistant cells, site-directed mutagenesis and photo-affinity labeling suggest that they compete with ATP binding via a novel allosteric mechanism.


Asunto(s)
Adenosina Trifosfato/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Concentración 50 Inhibidora , Modelos Moleculares , Estructura Molecular , Estructura Terciaria de Proteína
7.
Clin Cancer Res ; 14(23): 7588-92, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047083

RESUMEN

Cell cycle checkpoints have long been recognized as important nodes for regulating cell proliferation and maintaining genomic integrity. These checkpoints are often altered in cancer and represent promising points for therapeutic intervention. Until recently, direct targeting of the mitotic checkpoint has been an untapped area for cancer drug discovery. Regulation of the mitotic checkpoint is complex, but many of the critical players have been identified and functionally characterized. A substantial number of these proteins can be localized to the kinetochore, a structure located at the centromeric region of each mitotic chromosome. The kinetochore mediates chromosome attachment to spindle microtubules and subsequent chromosome movement. The mitotic checkpoint monitors microtubule attachment and chromosome position on the mitotic spindle, inhibiting progression into anaphase until proper attachment and metaphase positioning is achieved. Centromere-associated protein E is a kinesin microtubule motor protein that plays an essential role in integrating the mechanics of microtubule-chromosome interactions with mitotic checkpoint signaling, and has emerged as a novel target for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Cromosómicas no Histona/efectos de los fármacos , Proteínas Cromosómicas no Histona/metabolismo , Neoplasias/tratamiento farmacológico , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Genes cdc/efectos de los fármacos , Genes cdc/fisiología , Humanos , Mitosis/efectos de los fármacos , Mitosis/fisiología
8.
J Med Chem ; 50(20): 4939-52, 2007 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-17725339

RESUMEN

Kinesin spindle protein (KSP), an ATPase responsible for spindle pole separation during mitosis that is present only in proliferating cells, has become a novel and attractive anticancer target with potential for reduced side effects compared to currently available therapies. We report herein the discovery of the first known ATP-competitive inhibitors of KSP, which display a unique activity profile as compared to the known loop 5 (L5) allosteric KSP inhibitors that are currently under clinical evaluation. Optimization of this series led to the identification of biphenyl sulfamide 20, a potent KSP inhibitor with in vitro antiproliferative activity against human cells with either wild-type KSP (HCT116) or mutant KSP (HCT116 D130V). In a murine xenograft model with HCT116 D130V tumors, 20 showed significant antitumor activity following intraperitoneal dosing, providing in vivo proof-of-principle of the efficacy of an ATP-competitive KSP inhibitor versus tumors that are resistant to the other known KSP inhibitors.


Asunto(s)
Adenosina Trifosfato/metabolismo , Antineoplásicos/síntesis química , Compuestos de Bifenilo/síntesis química , Cinesinas/antagonistas & inhibidores , Sulfonamidas/síntesis química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacocinética , Compuestos de Bifenilo/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Cinesinas/genética , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Relación Estructura-Actividad , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología
9.
Cancer Res ; 64(9): 3276-80, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15126370

RESUMEN

Several members of the kinesin family of microtubule motor proteins play essential roles in mitotic spindle function and are potential targets for the discovery of novel antimitotic cancer therapies. KSP, also known as HsEg5, is a kinesin that plays an essential role in formation of a bipolar mitotic spindle and is required for cell cycle progression through mitosis. We identified a potent inhibitor of KSP, CK0106023, which causes mitotic arrest and growth inhibition in several human tumor cell lines. Here we show that CK0106023 is an allosteric inhibitor of KSP motor domain ATPase with a Ki of 12 nM. Among five kinesins tested, CK0106023 was specific for KSP. In tumor-bearing mice, CK0106023 exhibited antitumor activity comparable to or exceeding that of paclitaxel and caused the formation of monopolar mitotic figures identical to those produced in cultured cells. KSP was most abundant in proliferating human tissues and was absent from cultured postmitotic neurons. These findings are the first to demonstrate the feasibility of targeting mitotic kinesins for the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Cinesinas/antagonistas & inhibidores , Pirimidinas/farmacología , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/metabolismo , Animales , Femenino , Humanos , Cinesinas/metabolismo , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Clin Cancer Res ; 16(2): 566-76, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20068098

RESUMEN

PURPOSE: Ispinesib (SB-715992) is a potent inhibitor of kinesin spindle protein, a kinesin motor protein essential for the formation of a bipolar mitotic spindle and cell cycle progression through mitosis. Clinical studies of ispinesib have shown a 9% response rate in patients with locally advanced or metastatic breast cancer and a favorable safety profile without significant neurotoxicities, gastrointestinal toxicities, or hair loss. To better understand the potential of ispinesib in the treatment of breast cancer, we explored the activity of ispinesib alone and in combination with several therapies approved for the treatment of breast cancer. EXPERIMENTAL DESIGN: We measured the ispinesib sensitivity and pharmacodynamic response of breast cancer cell lines representative of various subtypes in vitro and as xenografts in vivo and tested the ability of ispinesib to enhance the antitumor activity of approved therapies. RESULTS: In vitro, ispinesib displayed broad antiproliferative activity against a panel of 53 breast cell lines. In vivo, ispinesib produced regressions in each of five breast cancer models and tumor-free survivors in three of these models. The effects of ispinesib treatment on pharmacodynamic markers of mitosis and apoptosis were examined in vitro and in vivo, revealing a greater increase in both mitotic and apoptotic markers in the MDA-MB-468 model than in the less sensitive BT-474 model. In vivo, ispinesib enhanced the antitumor activity of trastuzumab, lapatinib, doxorubicin, and capecitabine and exhibited activity comparable with paclitaxel and ixabepilone. CONCLUSIONS: These findings support further clinical exploration of kinesin spindle protein inhibitors for the treatment of breast cancer.


Asunto(s)
Benzamidas/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Cinesinas/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Quinazolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
ACS Med Chem Lett ; 1(1): 30-4, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24900171

RESUMEN

Inhibition of mitotic kinesins represents a novel approach for the discovery of a new generation of anti-mitotic cancer chemotherapeutics. We report here the discovery of the first potent and selective inhibitor of centromere-associated protein E (CENP-E) 3-chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide (GSK923295; 1), starting from a high-throughput screening hit, 3-chloro-4-isopropoxybenzoic acid 2. Compound 1 has demonstrated broad antitumor activity in vivo and is currently in human clinical trials.

13.
Biochemistry ; 47(11): 3576-85, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18290633

RESUMEN

KSP, also known as HsEg5, is a kinesin that plays an essential role in the formation of a bipolar mitotic spindle and is required for cell cycle progression through mitosis. Ispinesib is the first potent, highly specific small-molecule inhibitor of KSP tested for the treatment of human disease. This novel anticancer agent causes mitotic arrest and growth inhibition in several human tumor cell lines and is currently being tested in multiple phase II clinical trials. In this study we have used steady-state and pre-steady-state kinetic assays to define the mechanism of KSP inhibition by ispinesib. Our data show that ispinesib alters the ability of KSP to bind to microtubules and inhibits its movement by preventing the release of ADP without preventing the release of the KSP-ADP complex from the microtubule. This type of inhibition is consistent with the physiological effect of ispinesib on cells, which is to prevent KSP-driven mitotic spindle pole separation. A comparison of ispinesib to monastrol, another small-molecule inhibitor of KSP, reveals that both inhibitors share a common mode of inhibition.


Asunto(s)
Benzamidas/química , Cinesinas/antagonistas & inhibidores , Cinesinas/química , Quinazolinas/química , Adenosina Difosfato/antagonistas & inhibidores , Adenosina Difosfato/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfato/fisiología , Regulación Alostérica/genética , Benzamidas/metabolismo , Unión Competitiva/genética , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Microtúbulos/genética , Microtúbulos/metabolismo , Modelos Biológicos , Modelos Químicos , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Quinazolinas/metabolismo
14.
Biochemistry ; 46(11): 3494-502, 2007 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-17302440

RESUMEN

The Wiskott-Aldrich syndrome protein (WASP) and neural WASP (N-WASP) are key players in regulating actin cytoskeleton via the Arp2/3 complex. It has been widely reported that the WASP proteins are activated by Rho family small GTPase Cdc42 and that Rac1 acts through SCAR/WAVE proteins. However, a systematic study of the specificity of different GTPases for different Arp2/3 activators has not been conducted. In this study, we have expressed, purified, and characterized completely soluble, highly active, and autoinhibited full-length human WASP and N-WASP from mammalian cells. We show a novel N-WASP activation by Rho family small GTPase Rac1. This GTPase exclusively stimulates N-WASP and has no effects on WASP. Rac1 is a significantly more potent N-WASP activator than Cdc42. In contrast, Cdc42 is a more effective activator of WASP than N-WASP. Lipid vesicles containing PIP2 significantly improve actin nucleation by the Arp2/3 complex and N-WASP in the presence of Rac1 or Cdc42. PIP2 vesicles have no effect on WASP activity alone. Moreover, the inhibition of WASP-stimulated actin nucleation in the presence of Cdc42 and PIP2 vesicles has been observed. We found that adaptor proteins Nck1 or Nck2 are the most potent WASP and N-WASP activators with distinct effects on the WASP family members. Our in vitro data demonstrates differential regulation of full-length WASP and N-WASP by cellular activators that highlights fundamental differences of response at the protein-protein level.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/fisiología , Actinas/metabolismo , Proteínas Oncogénicas/fisiología , Fosfatidilinositol 4,5-Difosfato/fisiología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/fisiología , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Proteína de Unión al GTP cdc42/fisiología , Proteína de Unión al GTP rac1/fisiología , Proteínas Adaptadoras Transductoras de Señales , Humanos , Proteínas Recombinantes/biosíntesis , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA