Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Bioorg Med Chem Lett ; 24(6): 1466-71, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24582987

RESUMEN

This communication discusses the discovery of novel reverse tricyclic pyridones as inhibitors of Janus kinase 2 (JAK2). By using a kinase cross screening approach coupled with molecular modeling, a unique inhibitor-water interaction was discovered to impart excellent broad kinase selectivity. Improvements in intrinsic potency were achieved by utilizing a rapid library approach, while targeted structural changes to lower lipophilicity led to improved rat pharmacokinetics. This multi-pronged approach led to the identification of 31, which demonstrated encouraging rat pharmacokinetics, in vivo potency, and excellent off-target kinase selectivity.


Asunto(s)
Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Piridonas/química , Sulfonamidas/química , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Animales , Sitios de Unión , Evaluación Preclínica de Medicamentos , Semivida , Janus Quinasa 2/metabolismo , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Estructura Terciaria de Proteína , Piridonas/síntesis química , Piridonas/farmacocinética , Ratas , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética
2.
Bioorg Med Chem Lett ; 24(8): 1968-73, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24666646

RESUMEN

A series of carboxamide-substituted thiophenes demonstrating inhibition of JAK2 is described. Development of this chemical series began with the bioisosteric replacement of a urea substituent by a pyridyl ring. Issues of chemical and metabolic stability were solved using the results of both in vitro and in vivo studies, ultimately delivering compounds such as 24 and 25 that performed well in an acute PK/PD model measuring p-STAT5 inhibition.


Asunto(s)
Aminoimidazol Carboxamida/síntesis química , Aminoimidazol Carboxamida/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Tiofenos/síntesis química , Tiofenos/farmacología , Aminoimidazol Carboxamida/química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Microsomas/efectos de los fármacos , Microsomas/enzimología , Modelos Biológicos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Tiofenos/química
3.
Invest New Drugs ; 30(6): 2113-20, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22127459

RESUMEN

The poly-(ADP-ribose) polymerase (PARP) inhibitor, MK-4827, is a novel potent, orally bioavailable PARP-1 and PARP-2 inhibitor currently in phase I clinical trials for cancer treatment. No preclinical data currently exist on the combination of MK-4827 with radiotherapy. The current study examined combined treatment efficacy of MK-4827 and fractionated radiotherapy using a variety of human tumor xenografts of differing p53 status: Calu-6 (p53 null), A549 (p53 wild-type [wt]) and H-460 (p53 wt) lung cancers and triple negative MDA-MB-231 human breast carcinoma. To mimic clinical application of radiotherapy, fractionated radiation (2 Gy per fraction) schedules given once or twice daily for 1 to 2 weeks combined with MK-4827, 50 mg/kg once daily or 25 mg/kg twice daily, were used. MK-4827 was found to be highly and similarly effective in both radiation schedules but maximum radiation enhancement was observed when MK-4827 was given at a dose of 50 mg/kg once daily (EF = 2.2). MK-4827 radiosensitized all four tumors studied regardless of their p53 status. MK-4827 reduced PAR levels in tumors by 1 h after administration which persisted for up to 24 h. This long period of PARP inhibition potentially adds to the flexibility of design of future clinical trials. Thus, MK-4827 shows high potential to improve the efficacy of radiotherapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/terapia , Indazoles/administración & dosificación , Neoplasias Pulmonares/terapia , Piperidinas/administración & dosificación , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quimioradioterapia , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Neurochem ; 114(1): 191-202, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20403073

RESUMEN

NMDA receptors (NMDAR) contribute to neuronal development throughout the CNS. However, their mode(s) of activation preceding synaptic maturation is unclear, as they are not co-localized with alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate receptors (AMPARs) which normally provide sufficient depolarization to relieve voltage-dependent blockade by Mg(2+). We used cerebellar granule neurons (CGNs) cultured at a near-physiological KCl concentration to examine maturation-dependent changes in NMDAR responses. In contrast, most studies use KCl-supplemented medium to promote survival. At 2-4 days in vitro CGNs: (i) express developmental markers resembling the in vivo migratory phenotype; (ii) maintain a basal amount of calcium responsive element-binding protein phosphorylation that requires NMDARs and calcium/calmodulin-dependent kinases, but not AMPARs; (iii) exhibit NMDA-mediated Ca(2+) influx not effectively blocked by ambient Mg(2+) (0.75 mM) or AMPARs; (iv) maintain a more depolarized resting membrane potential and increased resistance compared to synaptically-connected CGNs. Moreover, migrating CGNs in explant cultures demonstrate NMDA-mediated Ca(2+) influx not effectively blocked by 0.75 mM Mg(2+), and NMDAR but not AMPAR antagonists slow migration. These data suggest the biophysical properties of immature CGNs render NMDARs less sensitive to Mg(2+) blockade, enhancing the likelihood of activation in the absence of AMPAR depolarization.


Asunto(s)
Cerebelo/metabolismo , Espacio Extracelular/metabolismo , Magnesio/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Calcio/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Movimiento Celular , Células Cultivadas , Cerebelo/citología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Potenciales de la Membrana , Fosforilación , Ratas , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/fisiología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Transducción de Señal , Sinapsis/fisiología
6.
J Biomol Screen ; 14(4): 404-11, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19403923

RESUMEN

c-Met is a receptor tyrosine kinase (RTK) with a critical role in many fundamental cellular processes, including cell proliferation and differentiation. Deregulated c-Met signaling has been implicated in both the initiation and progression of human cancers and therefore represents an attractive target for anticancer therapy. Monitoring the phosphorylation status of relevant tyrosine residues provides an important method of assessing c-Met kinase activity. This report describes a novel assay to monitor c-Met phosphorylation in cells using Amplified Luminescent Proximity Homogeneous Assay (AlphaScreen) technology. Using AlphaScreen, the authors were able to detect both global and site-specific phosphorylation of c-Met in transformed cell lines. Data obtained from the AlphaScreen assay were compared to data obtained from a high-content imaging (HCI) method developed in parallel to monitor c-Met phosphorylation at the single cell level. The AlphaScreen assay was miniaturized to a 384-well format with acceptable signal-to-background ratio (S/B) and Z' statistics and was employed to measure c-Met kinase activity in situ after treatment with potent c-Met-specific kinase inhibitors. The authors discuss the utility of quantifying endogenous cellular c-Met phosphorylation in lead optimization and how the modular design of the AlphaScreen assay allows its adaptation to measure cellular activity of other kinases.


Asunto(s)
Bioensayo/métodos , Imagenología Tridimensional/métodos , Proteínas Proto-Oncogénicas c-met/metabolismo , Recuento de Células , Línea Celular Tumoral , Humanos , Indicadores y Reactivos , Fosforilación , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Reproducibilidad de los Resultados
7.
Biochem Pharmacol ; 67(10): 1947-64, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15130771

RESUMEN

Cyclin-dependent kinases (CDKs) mediate proliferation and neuronal development, while aberrant CDK activity is associated with cancer and neurodegeneration. Consequently, pharmacologic inhibitors, such as 2,6,9-trisubstituted purines, which potently inhibit CDKs 1, 2, and 5, were developed to combat these pathologies. One agent, R-roscovitine (CYC202), has advanced to clinical trials as a potential cancer therapy. In primary neuronal cultures, these agents have been used to delineate the physiologic and pathologic functions of CDKs, and associated signaling pathways. Herein we demonstrate that three 2,6,9-trisubstituted purines: olomoucine, roscovitine, and purvalanol, used at concentrations ascribed by others to potently inhibit CDKs 1, 2, and 5, are powerful triggers of death in maturing cerebellar granule neurons, assessed by loss of mitochondrial reductive capacity and differential staining with fluorescent indicators of living/dead neurons. Based on several criteria, including delayed time course and establishment of an irreversible commitment point of death, pyknotic cell and nuclear morphology, and caspase-3 cleavage, the death process is apoptotic. However, pharmacological and biochemical data indicate that apoptosis is independent of CDK 1, 2, or 5 inhibition. This is based on the pattern of changes in c-jun mRNA, c-Jun protein, and Ca(2+)/cAMP response element binding protein (CREB) phosphorylation, and also, the ineffectiveness of structurally distinct CDK 1, 2, and 5 inhibitors butyrolactone-1 and PNU112445A to induce apoptosis. Collectively, our results, and those of others, indicate that the CDK regulation of transcription (CDKs 7 and 9) should be examined as a target of these agents, and as an indirect mediator of neuronal fate.


Asunto(s)
4-Butirolactona/análogos & derivados , Apoptosis , Cerebelo/citología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas , 4-Butirolactona/farmacología , Animales , Proteína Quinasa CDC2/metabolismo , Quinasas CDC2-CDC28/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Quinasa 5 Dependiente de la Ciclina , Quinasa 9 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Cinetina , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/citología , Fosforilación , Purinas/farmacología , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Roscovitina , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
8.
PLoS One ; 9(6): e98515, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24968263

RESUMEN

Aberrant kinase activation resulting from mutation, amplification, or translocation can drive growth and survival in a subset of human cancer. FGFR2 is amplified in breast and gastric cancer, and we report here the first characterization of FGFR2 gene amplification in colorectal cancer in the NCI-H716 colorectal cancer cell line. FGFR2 is highly expressed and activated in NCI-H716 cells, and FGFR selective small molecule inhibitors or FGFR2 shRNA strongly inhibited cell viability in vitro, indicating "addiction" of NCI-H716 cells to FGFR2. NCI-H716 growth in a xenograft model was also inhibited by an FGFR small molecule inhibitor. FGFR2 was required for activation of multiple downstream signaling proteins including AKT, ERK, S6RP and NFKB. Inhibition of downstream kinases such as AKT or ERK alone had modest effects on proliferation, whereas combined inhibition of AKT and ERK signaling resulted in a loss of viability similar to FGFR2 inhibition. We identified elevated FGFR2 expression in a small subset of primary colorectal cancer, however FGFR2 amplification was not observed. Although FGFR2 amplification is not common in primary colon cancer or lymph node and liver metastases, other subsets of colorectal cancer such as ascites, from which the NCI-H716 cell line was derived, have yet to be tested. These results suggest that emerging FGFR inhibitor therapeutics may have efficacy in a subset of colon cancer driven by FGFR2 amplification.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/genética , Amplificación de Genes , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Animales , Línea Celular Tumoral , Supervivencia Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
PLoS One ; 7(5): e37207, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22623993

RESUMEN

A high percentage of patients with the myeloproliferative disorder polycythemia vera (PV) harbor a Val617→Phe activating mutation in the Janus kinase 2 (JAK2) gene, and both cell culture and mouse models have established a functional role for this mutation in the development of this disease. We describe the properties of MRLB-11055, a highly potent inhibitor of both the WT and V617F forms of JAK2, that has therapeutic efficacy in erythropoietin (EPO)-driven and JAK2V617F-driven mouse models of PV. In cultured cells, MRLB-11055 blocked proliferation and induced apoptosis in a manner consistent with JAK2 pathway inhibition. MRLB-11055 effectively prevented EPO-induced STAT5 activation in the peripheral blood of acutely dosed mice, and could prevent EPO-induced splenomegaly and erythrocytosis in chronically dosed mice. In a bone marrow reconstituted JAK2V617F-luciferase murine PV model, MRLB-11055 rapidly reduced the burden of JAK2V617F-expressing cells from both the spleen and the bone marrow. Using real-time in vivo imaging, we examined the kinetics of disease regression and resurgence, enabling the development of an intermittent dosing schedule that achieved significant reductions in both erythroid and myeloid populations with minimal impact on lymphoid cells. Our studies provide a rationale for the use of non-continuous treatment to provide optimal therapy for PV patients.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Policitemia Vera/tratamiento farmacológico , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/uso terapéutico , Eritropoyetina/metabolismo , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT5/metabolismo
10.
PLoS One ; 4(9): e7286, 2009 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-19789710

RESUMEN

Splenic enlargement (splenomegaly) develops in numerous disease states, although a specific pathogenic role for the spleen has rarely been described. In polycythemia vera (PV), an activating mutation in Janus kinase 2 (JAK2(V617)) induces splenomegaly and an increase in hematocrit. Splenectomy is sparingly performed in patients with PV, however, due to surgical complications. Thus, the role of the spleen in the pathogenesis of human PV remains unknown. We specifically tested the role of the spleen in the pathogenesis of PV by performing either sham (SH) or splenectomy (SPL) surgeries in a murine model of JAK2(V617F)-driven PV. Compared to SH-operated mice, which rapidly develop high hematocrits after JAK2(V617F) transplantation, SPL mice completely fail to develop this phenotype. Disease burden (JAK2(V617)) is equivalent in the bone marrow of SH and SPL mice, however, and both groups develop fibrosis and osteosclerosis. If SPL is performed after PV is established, hematocrit rapidly declines to normal even though myelofibrosis and osteosclerosis again develop independently in the bone marrow. In contrast, SPL only blunts hematocrit elevation in secondary, erythropoietin-induced polycythemia. We conclude that the spleen is required for an elevated hematocrit in murine, JAK2(V617F)-driven PV, and propose that this phenotype of PV may require a specific interaction between mutant cells and the spleen.


Asunto(s)
Hematócrito , Janus Quinasa 2/genética , Policitemia Vera/sangre , Policitemia Vera/cirugía , Esplenectomía/métodos , Alelos , Animales , Médula Ósea/metabolismo , Trasplante de Médula Ósea , Eritropoyetina/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación , Fenotipo , Bazo/metabolismo
11.
Biochem Pharmacol ; 78(4): 382-9, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19413997

RESUMEN

Polycythemia vera (PV) is a myeloproliferative disorder characterized by increased red cell mass and splenomegaly in the absence of secondary causes [Tefferi A., Spivak J.L., Polycythemia vera: scientific advances and current practice. Semin Hematol 2005;42(4):206-20.]. Recently, several laboratories have discovered that the vast majority of patients with PV carry a single, activating mutation (V617F) in the pseudokinase domain of Janus kinase 2 (Jak2) [Zhao R, Xing S, Li Z, Fu X, Li Q, Krantz SB, et al., Identification of an acquired JAK2 mutation in polycythemia vera. J Biol Chem 2005;280(24):22788-92; James C, Ugo V, Le Couédic JP, Staerk J, Delhommeau F, Lacout C, et al., A unique clonal JAK2 mutation leading to constitutive signalling causes polycythemia vera. Nature 2005;434(7037):1144-8; Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, et al., A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005;352(17):1779-90; Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al., Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005;7(4):387-97.]. This discovery has spurred interest in developing therapies for PV via inhibition of Jak2. We induced polycythemia in mice by administering high dose recombinant erythropoietin (Epo) and determined that administration recapitulates almost all of the major and minor diagnostic features of human PV. We then tested a selective, small molecule inhibitor of Jak2 (Jak2i) and showed that this treatment prevents polycythemia. This prevention of polycythemia was accompanied by lower hematocrits, reduced spleen sizes and reductions in Stat5 phosphorylation (pStat5). Surprisingly, Epo rapidly (<1h) induces mobilization of activated erythroid precursors into the blood, thus allowing drug-response relationships to guide discovery. We conclude that inhibition of Jak2 prevents polycythemia in mice, and furthermore present this model as an efficient tool for the discovery of drugs that effectively treat human PV.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Janus Quinasa 2/antagonistas & inhibidores , Policitemia Vera/fisiopatología , Policitemia/prevención & control , Piridonas/uso terapéutico , Animales , Proliferación Celular , Inhibidores Enzimáticos/farmacología , Células Precursoras Eritroides , Humanos , Janus Quinasa 2/metabolismo , Ratones , Fosforilación , Mielofibrosis Primaria/fisiopatología , Proteínas Tirosina Quinasas/metabolismo , Piridonas/síntesis química , Piridonas/química , Transducción de Señal , Trombocitemia Esencial , Células Tumorales Cultivadas
12.
J Neurochem ; 81(2): 314-24, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12064479

RESUMEN

CaMKIV is enriched in neuronal nuclei and mediates Ca2+-dependent survival via transcription factor phosphorylation. Cultured cerebellar granule neurons were used to examine whether distinct modes of Ca2+ signaling differentially modulate CaMKIV expression and function. For long-term survival, these neurons require 25 mm KCl or NMDA, which stimulates Ca2+ entry through voltage-sensitive Ca2+ channels or NMDA receptors (NRs). Lower levels of Ca2+ entry through NRs support survival of a neuronal subpopulation grown in 5 mm KCl media. Several effects were demonstrated: (i) sustained exposure to 25 mM KCl or 140 microM NMDA produced CaMKIV down-regulation, compared to 5 mM KCl cultures; (ii) CaMKIV down-regulation was attenuated by nifedipine, APV and CaM kinase inhibitors, indicating that it is Ca2+ dependent and reversible; (iii) down-regulation was both selective for nuclear substrates and calpain-mediated; (iv) proteolysis was exacerbated by leptomycin B, a nuclear export inhibitor. Although CaMKIV proteolysis by trophic agents seems paradoxical in light of evidence supporting its critical role in survival, the CaMKIV/CREB signal transduction pathway was preserved, as assessed by CaM kinase-mediated CREB phosphorylation, and the ability of CaM kinase inhibitors to interfere with KCl-mediated survival. We hypothesize that limited calpain-mediated proteolysis of CaMKIV is a negative feedback response to the sustained activation of a Ca2+ and CaMKIV signaling pathway by these agents.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calpaína/metabolismo , N-Metilaspartato/farmacología , Neuronas/metabolismo , Cloruro de Potasio/farmacología , Receptores Citoplasmáticos y Nucleares , Transporte Activo de Núcleo Celular/efectos de los fármacos , Adaptación Fisiológica/fisiología , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cerebelo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ácidos Grasos Insaturados/farmacología , Carioferinas/antagonistas & inhibidores , Neuronas/citología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA