Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Med Chem Lett ; 10(9): 1302-1308, 2019 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-31531201

RESUMEN

KRAS regulates many cellular processes including proliferation, survival, and differentiation. Point mutants of KRAS have long been known to be molecular drivers of cancer. KRAS p.G12C, which occurs in approximately 14% of lung adenocarcinomas, 3-5% of colorectal cancers, and low levels in other solid tumors, represents an attractive therapeutic target for covalent inhibitors. Herein, we disclose the discovery of a class of novel, potent, and selective covalent inhibitors of KRASG12C identified through a custom library synthesis and screening platform called Chemotype Evolution and structure-based design. Identification of a hidden surface groove bordered by H95/Y96/Q99 side chains was key to the optimization of this class of molecules. Best-in-series exemplars exhibit a rapid covalent reaction with cysteine 12 of GDP-KRASG12C with submicromolar inhibition of downstream signaling in a KRASG12C-specific manner.

2.
Bioorg Med Chem Lett ; 21(10): 3078-83, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21459573

RESUMEN

We report the use of a fragment-based lead discovery method, Tethering with extenders, to discover a pyridinone fragment that binds in an adaptive site of the protein PDK1. With subsequent medicinal chemistry, this led to the discovery of a potent and highly selective inhibitor of PDK1, which binds in the 'DFG-out' conformation.


Asunto(s)
Diseño de Fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Cristalografía por Rayos X , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Concentración 50 Inhibidora , Modelos Biológicos , Estructura Molecular , Piridonas/química , Piridonas/farmacología , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
3.
Cancer Chemother Pharmacol ; 65(4): 707-17, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19649632

RESUMEN

PURPOSE: The Aurora family of serine/threonine kinases (Aurora-A, Aurora-B, and Aurora-C) plays a key role in cells orderly progression through mitosis. Elevated expression levels of Aurora kinases have been detected in a high percentage of melanoma, colon, breast, ovarian, gastric, and pancreatic tumors. We characterized the biological and pharmacological properties of SNS-314, an ATP-competitive, selective, and potent inhibitor of Aurora kinases. METHODS: We studied the biochemical potency and selectivity of SNS-314 to inhibit Aurora kinases A, B, and C. The inhibition of cellular proliferation induced by SNS-314 was evaluated in a broad range of tumor cell lines and correlated to inhibition of histone H3 phosphorylation, inhibition of cell-cycle progression, increase in nuclear content and cell size, loss of viability, and induction of apoptosis. The dose and administration schedule of SNS-314 was optimized for in vivo efficacy in mouse xenograft models of human cancer. RESULTS: In the HCT116 human colon cancer xenograft model, administration of 50 and 100 mg/kg SNS-314 led to dose-dependent inhibition of histone H3 phosphorylation for at least 10 h, indicating effective Aurora-B inhibition in vivo. HCT116 tumors from animals treated with SNS-314 showed potent and sustained responses including reduction of phosphorylated histone H3 levels, increased caspase-3 and appearance of increased nuclear size. The compound showed significant tumor growth inhibition in a dose-dependent manner under a variety of dosing schedules including weekly, bi-weekly, and 5 days on/9 days off. CONCLUSIONS: SNS-314 is a potent small-molecule inhibitor of Aurora kinases developed as a novel anti-cancer therapeutic agent for the treatment of diverse human malignancies.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/prevención & control , Compuestos de Fenilurea/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Tiazoles/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Aurora Quinasa A , Aurora Quinasa B , Aurora Quinasa C , Aurora Quinasas , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Femenino , Células HCT116 , Células HT29 , Células HeLa , Histonas/metabolismo , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias/metabolismo , Neoplasias/patología , Compuestos de Fenilurea/química , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Tiazoles/química , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Protein Expr Purif ; 54(1): 139-46, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17434748

RESUMEN

Aurora kinases have recently become some of the most intensely pursued oncology targets for the design of small-molecule inhibitors. Most of the active Aurora-A protein variants are currently being expressed from baculoviruses in insect cells, while catalytically impaired proteins can also be generated in and purified from Escherichia coli. In this study we present a method of expressing large quantities of active mouse Aurora-A kinase domain as an N-terminal glutathione-S-transferase fusion protein in bacteria and outline a simple purification method that produces greater than 99% pure protein samples suitable for enzymatic assays and X-ray crystallography. The methods described in this report simplify mouse Aurora-A expression and purification, and may aid in the production of other difficult kinases in prokaryotes.


Asunto(s)
Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Animales , Aurora Quinasa A , Aurora Quinasas , Cristalización , Cristalografía por Rayos X , Escherichia coli/genética , Glutatión Transferasa/genética , Ratones , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes/química
5.
Biochemistry ; 44(21): 7704-12, 2005 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-15909985

RESUMEN

The fluorogenic reagent 4-(aminosulfonyl)-7-fluoro-2,1,3-benzoxadiazole (ABDF) attenuates the functional activity of the protein tyrosine phosphatase PTP1B by reacting selectively with a single cysteine residue, leaving other cysteines in the protein unmodified. This modification reduces Vmax without substantially affecting substrate binding (Km), indicative of an allosteric mode of inhibition. Consistent with this, the cysteine residue modified by ABDF, Cys 121, lies outside the catalytic site but makes interactions with residues that contact His 214, which has been shown to be important for catalysis. Cys 121 is highly conserved among phosphatases, and ABDF also inhibits TC-PTP and LAR. These findings illustrate that targeting cysteine residues outside catalytic sites may be exploited in allosterically regulating enzymes. Moreover, these results suggest a new strategy for inhibiting a promising diabetes target.


Asunto(s)
Cisteína/metabolismo , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , Regulación Alostérica , Sitio Alostérico , Animales , Células CHO , Catálisis , Cricetinae , Inhibidores Enzimáticos/química , Polarización de Fluorescencia , Colorantes Fluorescentes/química , Humanos , Insulina/fisiología , Cinética , Oxadiazoles/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/química , Transducción de Señal/fisiología , Espectrometría de Masa por Ionización de Electrospray
6.
Curr Opin Chem Biol ; 8(4): 399-406, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15288250

RESUMEN

Rapid progress in genomics and proteomics has provided a wealth of new targets for the pharmaceutical industry, even as many older targets still remain challenging for small-molecule drug discovery. Fragment-based lead discovery, in which leads are built progressively by expanding or combining small fragments, is a rapidly growing field that offers potential advantages over traditional lead-discovery processes. However, identifying and assembling the fragments themselves can be challenging. Here, we review the concept of site-directed ligand discovery, in which a covalent bond is used to stabilize the interaction between a low-affinity fragment and a target protein. We also describe how this technique can facilitate fragment-based lead discovery and help overcome some of the limitations of traditional screening methods.


Asunto(s)
Diseño de Fármacos , Fragmentos de Péptidos/metabolismo , Proteínas/metabolismo , Sitios de Unión/fisiología , Ligandos , Fragmentos de Péptidos/química , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Proteínas/química
7.
Nat Struct Mol Biol ; 11(8): 730-7, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15258570

RESUMEN

Obesity and type II diabetes are closely linked metabolic syndromes that afflict >100 million people worldwide. Although protein tyrosine phosphatase 1B (PTP1B) has emerged as a promising target for the treatment of both syndromes, the discovery of pharmaceutically acceptable inhibitors that bind at the active site remains a substantial challenge. Here we describe the discovery of an allosteric site in PTP1B. Crystal structures of PTP1B in complex with allosteric inhibitors reveal a novel site located approximately 20 A from the catalytic site. We show that allosteric inhibitors prevent formation of the active form of the enzyme by blocking mobility of the catalytic loop, thereby exploiting a general mechanism used by tyrosine phosphatases. Notably, these inhibitors exhibit selectivity for PTP1B and enhance insulin signaling in cells. Allosteric inhibition is a promising strategy for targeting PTP1B and constitutes a mechanism that may be applicable to other tyrosine phosphatases.


Asunto(s)
Proteínas Tirosina Fosfatasas/química , Sitio Alostérico , Animales , Sitios de Unión , Unión Competitiva , Células CHO , Catálisis , Dominio Catalítico , Clonación Molecular , Cricetinae , Cristalografía por Rayos X , ADN/química , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Cinética , Ligandos , Modelos Químicos , Modelos Moleculares , Obesidad , Monoéster Fosfórico Hidrolasas/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Factores de Tiempo , Transfección , Tirosina/química
8.
Brain Res Mol Brain Res ; 124(2): 165-77, 2004 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-15135225

RESUMEN

The molecular cues that regulate neurite morphology within the target environment are key to the formation of complex neural circuitry. During development of the ponto-cerebellar projection, pontine fibers sprout and form elaborate arbors within the inner cerebellar layer prior to arrival of their target cells, the cerebellar granule neurons. Here, we describe the biochemical fractionation of two granule neuron-derived factors that stimulate elaboration of pontine neurites. These factors were identified using a dissociated pontine bioassay and biochemically fractionated from granule cell (GC) conditioned medium (GCCM). One of the factors, STIM1, is a protein with a molecular weight greater than 30 kDa that is distinct from known neurotrophins. The other, STIM2, is a small, protease-resistant molecule with an estimated molecular weight below 1 kDa. We show that these factors stimulate pontine neurite elongation both independently and cooperatively and thus may contribute to the formation of elaborate pontine arbors within the cerebellar cortex.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Factores de Crecimiento Nervioso/metabolismo , Vías Nerviosas/crecimiento & desarrollo , Neuritas/metabolismo , Puente/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Señales (Psicología) , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/metabolismo , Ratones , Modelos Biológicos , Peso Molecular , Factores de Crecimiento Nervioso/aislamiento & purificación , Factores de Crecimiento Nervioso/farmacología , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuritas/efectos de los fármacos , Puente/citología , Puente/metabolismo , Transducción de Señal/fisiología
9.
J Am Chem Soc ; 125(19): 5602-3, 2003 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-12733877

RESUMEN

Protein tyrosine phosphatases play important roles in many signaling cascades involved in human disease. The identification of druglike inhibitors for these targets is a major challenge, and the discovery of suitable phosphotyrosine (pY) mimetics remains one of the key difficulties. Here we describe an extension of tethering technology, "breakaway tethering", which is ideally suited for discovering such new chemical entities. The approach involves first irreversibly modifying a protein with an extender that contains both a masked thiol and a known pY mimetic. The extender is then cleaved to release the pY mimetic, unmasking the thiol. The resulting protein is screened against a library of disulfide-containing small molecule fragments; any molecules with inherent affinity for the pY binding site will preferentially form disulfides with the extender, allowing for their identification by mass spectrometry. The ability to start from a known substrate mimimizes perturbation of protein structure and increases the opportunity to probe the active site using tethering. We applied this approach to the anti-diabetic protein PTP1B to discover a pY mimetic which belongs to a new molecular class and which binds in a novel fashion.


Asunto(s)
Materiales Biomiméticos/química , Fosfotirosina/química , Proteínas Tirosina Fosfatasas/química , Sitios de Unión , Materiales Biomiméticos/metabolismo , Cristalografía por Rayos X , Cisteína/química , Modelos Moleculares , Ácido Oxálico/química , Fosfotirosina/metabolismo , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...