Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
2.
Bioorg Med Chem Lett ; 19(1): 226-9, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19019675

RESUMEN

An approach and preliminary results for utilizing legacy MEK inhibitors as templates for a reiterative structural based design and synthesis of novel, type III NCKIs (non-classical kinase inhibitors) is described. Evidence is provided that the MEK-pocket or pockets closely related to it may exist in kinases other than MEK.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/síntesis química , Dominio Catalítico , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
3.
Nat Struct Mol Biol ; 11(12): 1192-7, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15543157

RESUMEN

MEK1 and MEK2 are closely related, dual-specificity tyrosine/threonine protein kinases found in the Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) signaling pathway. Approximately 30% of all human cancers have a constitutively activated MAPK pathway, and constitutive activation of MEK1 results in cellular transformation. Here we present the X-ray structures of human MEK1 and MEK2, each determined as a ternary complex with MgATP and an inhibitor to a resolution of 2.4 A and 3.2 A, respectively. The structures reveal that MEK1 and MEK2 each have a unique inhibitor-binding pocket adjacent to the MgATP-binding site. The presence of the potent inhibitor induces several conformational changes in the unphosphorylated MEK1 and MEK2 enzymes that lock them into a closed but catalytically inactive species. Thus, the structures reported here reveal a novel, noncompetitive mechanism for protein kinase inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , MAP Quinasa Quinasa 1/química , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/química , MAP Quinasa Quinasa 2/metabolismo , Sitios de Unión , Secuencia Conservada , Dimerización , Inhibidores Enzimáticos/química , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Modelos Moleculares , Estructura Molecular , Estructura Cuaternaria de Proteína , Homología Estructural de Proteína
4.
Mol Cancer Ther ; 7(7): 1880-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18606718

RESUMEN

Signaling through the erbB receptor family of tyrosine kinases contributes to the proliferation, differentiation, migration, and survival of a variety of cell types. Abnormalities in members of this receptor family have been shown to play a role in oncogenesis, thus making them attractive targets for anticancer treatments. PF-00299804 is a second-generation irreversible pan-erbB receptor tyrosine kinase inhibitor currently in phase I clinical trials. PF-00299804 is believed to irreversibly inhibit erbB tyrosine kinase activity through binding at the ATP site and covalent modification of nucleophilic cysteine residues in the catalytic domains of erbB family members. Oral administration of PF-00299804 causes significant antitumor activity, including marked tumor regressions in a variety of human tumor xenograft models that express and/or overexpress erbB family members or contain the double mutation (L858R/T790M) in erbB1 (EGFR) associated with resistance to gefitinib and erlotinib. Furthermore, PF-00299804 shows exceptional distribution to human tumor xenografts and excellent pharmacokinetic properties across species.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Quinazolinonas/farmacología , Quinazolinonas/farmacocinética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Ratones SCID , Mutación/genética , Fosforilación/efectos de los fármacos , Especificidad de la Especie
5.
Bioorg Med Chem Lett ; 18(23): 6171-4, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18951019

RESUMEN

This paper reports a second generation MEK inhibitor. The previously reported potent and efficacious MEK inhibitor, PD-184352 (CI-1040), contains an integral hydroxamate moiety. This compound suffered from less than ideal solubility and metabolic stability. An oxadiazole moiety behaves as a bioisostere for the hydroxamate group, leading to a more metabolically stable and efficacious MEK inhibitor.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Benzamidas/farmacología , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Oxadiazoles/síntesis química , Oxadiazoles/farmacología , Antineoplásicos/química , Benzamidas/química , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/tratamiento farmacológico , Técnicas Químicas Combinatorias , Ensayos de Selección de Medicamentos Antitumorales , Ésteres , Humanos , Ácidos Hidroxámicos/química , Microsomas Hepáticos/efectos de los fármacos , Estructura Molecular , Oxadiazoles/química , Relación Estructura-Actividad
6.
Bioorg Med Chem Lett ; 18(24): 6501-4, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18952427

RESUMEN

A novel series of benzhydroxamate esters derived from their precursor anthranilic acids have been prepared and have been identified as potent MEK inhibitors. 2-(2-Chloro-4-iodo-phenylamino)-N-cyclopropylmethoxy-3,4-difluoro-benzamide, CI-1040, was the first MEK inhibitor to demonstrate in vivo activity in preclinical animal models and subsequently became the first MEK inhibitor to enter clinical trial. CI-1040 suffered however from poor exposure due to its poor solubility and rapid clearance, and as a result, development of the compound was terminated. Optimization of the diphenylamine core and modification of the hydroxamate side chain for cell potency, solubility, and exposure with oral delivery resulted in the discovery of the clinical candidate N-(2,3-dihydroxy-propoxy)-3,4-difluoro-2-(2-fluoro-4-iodo-phenylamino)-benzamide PD 0325901.


Asunto(s)
Benzamidas/síntesis química , Difenilamina/análogos & derivados , Inhibidores Enzimáticos/síntesis química , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Animales , Benzamidas/farmacología , Benzoatos/química , Línea Celular Tumoral , Química Farmacéutica/métodos , Difenilamina/síntesis química , Difenilamina/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Ácidos Hidroxámicos/química , Concentración 50 Inhibidora , Ratones , Trasplante de Neoplasias , Solubilidad , ortoaminobenzoatos/química
7.
J Med Chem ; 50(21): 5090-102, 2007 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17880056

RESUMEN

A new series of MEK1 inhibitors, the 4-anilino-5-carboxamido-2-pyridones, were designed and synthesized using a combination of medicinal chemistry, computational chemistry, and structural elucidation. The effect of variation in the carboxamide side chain, substitution on the pyridone nitrogen, and replacement of the 4'-iodide were all investigated. This study afforded several compounds which were either equipotent or more potent than the clinical candidate CI-1040 (1) in an isolated enzyme assay, as well as murine colon carcinoma (C26) cells, as measured by suppression of phosphorylated ERK substrate. Most notably, pyridone 27 was found to be more potent than 1 in vitro and produced a 100% response rate at a lower dose than 1, when tested for in vivo efficacy in animals bearing C26 tumors.


Asunto(s)
Amidas/síntesis química , Compuestos de Anilina/síntesis química , Antineoplásicos/síntesis química , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Piridonas/síntesis química , Amidas/química , Amidas/farmacología , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Benzamidas/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , MAP Quinasa Quinasa 1/química , MAP Quinasa Quinasa 2/química , Masculino , Ratones , Modelos Moleculares , Trasplante de Neoplasias , Fosforilación , Piridonas/química , Piridonas/farmacología , Ratas , Relación Estructura-Actividad
8.
J Med Chem ; 59(17): 8103-24, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27491023

RESUMEN

Structure-activity relationships for inhibition of erbB1, erbB2, and erbB4 were determined for a series of quinazoline- and pyrido[3,4-d]pyrimidine-based analogues of the irreversible pan-erbB inhibitor, canertinib. Cyclic amine bearing crotonamides were determined to provide rapid inhibition of cellular erbB1 autophosphorylation and good metabolic stability in liver microsome and hepatocyte assays. The influence of 4-anilino substitution on pan-erbB inhibitory potency was investigated. Several anilines were identified as providing potent, reversible pan-erbB inhibition. Optimum 4- and 6-substituents with known 7-substituents provided preferred irreversible inhibitors for pharmacodynamic testing in vivo. Quinazoline 54 and pyrido[3,4-d]pyrimidine 71 were identified as clearly superior to canertinib. Both compounds possess a piperidinyl crotonamide Michael acceptor and a 3-chloro-4-fluoroaniline, indicating these as optimized 6- and 4-substituents, respectively. Pharmacokinetic comparison of compounds 54 and 71 across three species selected compound 54 as the preferred candidate. Compound 54 (PF-00299804) has been assigned the nomenclature of dacomitinib and is currently under clinical evaluation.


Asunto(s)
Antineoplásicos/química , Receptores ErbB/antagonistas & inhibidores , Morfolinas/química , Piridinas/química , Pirimidinas/química , Quinazolinas/química , Quinazolinonas/química , Administración Oral , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Perros , Xenoinjertos , Humanos , Inyecciones Intravenosas , Macaca fascicularis , Masculino , Ratones Desnudos , Morfolinas/síntesis química , Morfolinas/farmacocinética , Morfolinas/farmacología , Trasplante de Neoplasias , Fosforilación , Piridinas/síntesis química , Piridinas/farmacocinética , Piridinas/farmacología , Pirimidinas/síntesis química , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Quinazolinas/síntesis química , Quinazolinas/farmacocinética , Quinazolinas/farmacología , Quinazolinonas/síntesis química , Quinazolinonas/farmacocinética , Quinazolinonas/farmacología , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad
9.
Chem Biol Drug Des ; 74(6): 547-59, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19843080

RESUMEN

The design, synthesis and utility of fluorescence probes that bind to the DFG-out conformation of p38alpha kinase are described. Probes that demonstrate good affinity for p38alpha, have been identified and one of the probes, PF-04438255, has been successfully used in an high throughput screening (HTS) assay to identify two novel non-classical p38alpha inhibitors. In addition, a cascade activity assay was utilized to validate the selective binding of these non-classical kinase inhibitors to the unactive form of the enzyme.


Asunto(s)
Colorantes Fluorescentes/síntesis química , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacología , Ensayos Analíticos de Alto Rendimiento , Cinética , Proteína Quinasa 14 Activada por Mitógenos/química , Naftalenos/química , Naftalenos/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/química , Pirazoles/farmacología , Relación Estructura-Actividad
10.
Arthritis Rheum ; 56(10): 3347-57, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17907188

RESUMEN

OBJECTIVE: To evaluate the role of the MEK/ERK MAP kinase pathway in murine collagen-induced arthritis (CIA) using the selective MEK inhibitor PD184352. We examined the effects of the inhibitor in cytokine-stimulated synovial fibroblasts and in cytokine-induced arthritis in rabbits to investigate its antiinflammatory mechanisms. METHODS: Murine CIA was used to assess the effects of the selective MEK inhibitor on paw edema, clinical scores, weight loss, histopathologic features, and joint levels of p-ERK. Western blotting and immunohistochemistry techniques were used to assess p-ERK in human and rabbit synovial fibroblasts and synovial tissue from rheumatoid arthritis (RA) patients. Interleukin-1alpha (IL-1alpha)-stimulated stromelysin production in rabbit synovial fibroblasts was assessed by enzyme-linked immunosorbent assay. A rabbit IL-1alpha-induced arthritis model was used to assess the effects of the inhibitor on IL-1alpha-induced MEK activity, stromelysin production, and cartilage degradation. RESULTS: In the CIA model, PD184352 inhibited paw edema and clinical arthritis scores in a dose-dependent manner. Disease-induced weight loss and histopathologic changes were also significantly improved by treatment. Inhibition of disease-induced p-ERK levels in the joints was seen with the inhibitor. Levels of p-ERK in the synovium were higher in RA patients than in normal individuals. PD184352 reduced IL-1alpha-induced p-ERK levels in human RA synovial fibroblasts. The production of p-ERK and stromelysin was also inhibited in IL-1alpha-stimulated rabbit synovial fibroblasts. We observed IL-1alpha-induced p-ERK in the synovial lining, subsynovial vasculature, and articular chondrocytes. IL-1alpha-induced stromelysin production and proteoglycan loss from the articular cartilage were reduced by PD184352. CONCLUSION: These data demonstrate the inhibition of murine CIA by PD184352, support the hypothesis that antiinflammatory activity contributes to the mechanism of action of the inhibitor, and suggest that a selective inhibitor may effectively treat RA and other inflammatory disorders.


Asunto(s)
Artritis Reumatoide/enzimología , Benzamidas/farmacología , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/efectos de los fármacos , Animales , Artritis Reumatoide/tratamiento farmacológico , Western Blotting , Cartílago Articular/efectos de los fármacos , Cartílago Articular/enzimología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Inmunohistoquímica , Técnicas In Vitro , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Ratones Endogámicos DBA , Conejos
11.
Bioorg Med Chem Lett ; 17(16): 4599-603, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17562362

RESUMEN

It has been hypothesized that peripherally restricted NMDA receptor antagonists may be effective analgesics for osteoarthritis pain. A class of novel quinoxalinedione atropisomers, first discovered for an NMDA receptor antagonist program for the treatment of stroke, was evaluated and further optimized with the goal of finding peripherally restricted NMDA receptor antagonists.


Asunto(s)
Analgésicos/química , Analgésicos/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sulfonamidas/química , Sulfonamidas/farmacología , Animales , Área Bajo la Curva , Sitios de Unión , Modelos Moleculares , Estructura Molecular , Dolor/tratamiento farmacológico , Unión Proteica , Ratas , Relación Estructura-Actividad , Sulfonamidas/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA