Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 59(11): 5221-37, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27167172

RESUMEN

Multiparameter optimization of a series of 5-((4-aminopyridin-2-yl)amino)pyrazine-2-carbonitriles resulted in the identification of a potent and selective oral CHK1 preclinical development candidate with in vivo efficacy as a potentiator of deoxyribonucleic acid (DNA) damaging chemotherapy and as a single agent. Cellular mechanism of action assays were used to give an integrated assessment of compound selectivity during optimization resulting in a highly CHK1 selective adenosine triphosphate (ATP) competitive inhibitor. A single substituent vector directed away from the CHK1 kinase active site was unexpectedly found to drive the selective cellular efficacy of the compounds. Both CHK1 potency and off-target human ether-a-go-go-related gene (hERG) ion channel inhibition were dependent on lipophilicity and basicity in this series. Optimization of CHK1 cellular potency and in vivo pharmacokinetic-pharmacodynamic (PK-PD) properties gave a compound with low predicted doses and exposures in humans which mitigated the residual weak in vitro hERG inhibition.


Asunto(s)
4-Aminopiridina/análogos & derivados , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , 4-Aminopiridina/síntesis química , 4-Aminopiridina/química , 4-Aminopiridina/farmacología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirazinas/síntesis química , Pirazinas/química , Relación Estructura-Actividad
2.
Oncotarget ; 7(3): 2329-42, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26295308

RESUMEN

CCT245737 is the first orally active, clinical development candidate CHK1 inhibitor to be described. The IC50 was 1.4 nM against CHK1 enzyme and it exhibited>1,000-fold selectivity against CHK2 and CDK1. CCT245737 potently inhibited cellular CHK1 activity (IC50 30-220 nM) and enhanced gemcitabine and SN38 cytotoxicity in multiple human tumor cell lines and human tumor xenograft models. Mouse oral bioavailability was complete (100%) with extensive tumor exposure. Genotoxic-induced CHK1 activity (pS296 CHK1) and cell cycle arrest (pY15 CDK1) were inhibited both in vitro and in human tumor xenografts by CCT245737, causing increased DNA damage and apoptosis. Uniquely, we show CCT245737 enhanced gemcitabine antitumor activity to a greater degree than for higher doses of either agent alone, without increasing toxicity, indicating a true therapeutic advantage for this combination. Furthermore, development of a novel ELISA assay for pS296 CHK1 autophosphorylation, allowed the quantitative measurement of target inhibition in a RAS mutant human tumor xenograft of NSCLC at efficacious doses of CCT245737. Finally, CCT245737 also showed significant single-agent activity against a MYC-driven mouse model of B-cell lymphoma. In conclusion, CCT245737 is a new CHK1 inhibitor clinical development candidate scheduled for a first in man Phase I clinical trial, that will use the novel pS296 CHK1 ELISA to monitor target inhibition.


Asunto(s)
4-Aminopiridina/análogos & derivados , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Linfoma de Células B/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Pirazinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , 4-Aminopiridina/farmacocinética , 4-Aminopiridina/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Proteína Quinasa CDC2 , Camptotecina/análogos & derivados , Camptotecina/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quinasa de Punto de Control 2/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Daño del ADN/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sinergismo Farmacológico , Células HT29 , Humanos , Irinotecán , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Pirazinas/farmacocinética , Gemcitabina
3.
Oncotarget ; 4(10): 1647-61, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24072592

RESUMEN

The ribosomal P70 S6 kinases play a crucial role in PI3K/mTOR regulated signalling pathways and are therefore potential targets for the treatment of a variety of diseases including diabetes and cancer. In this study we describe the identification of three series of chemically distinct S6K1 inhibitors. In addition, we report a novel PKA-S6K1 chimeric protein with five mutations in or near its ATP-binding site, which was used to determine the binding mode of two of the three inhibitor series, and provided a robust system to aid the optimisation of the oxadiazole-substituted benzimidazole inhibitor series. We show that the resulting oxadiazole-substituted aza-benzimidazole is a potent and ligand efficient S6 kinase inhibitor, which blocks the phosphorylation of RPS6 at Ser235/236 in TSC negative HCV29 human bladder cancer cells by inhibiting S6 kinase activity and thus provides a useful tool compound to investigate the function of S6 kinases.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diseño de Fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Imidazoles/química , Imidazoles/farmacología , Modelos Moleculares , Fosforilación , Inhibidores de Proteínas Quinasas/química , Proteínas Recombinantes de Fusión/química , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/enzimología
4.
PLoS One ; 8(6): e65689, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23776527

RESUMEN

Checkpoint kinase 2 (CHK2) is an important serine/threonine kinase in the cellular response to DNA damage. A fragment-based screening campaign using a combination of a high-concentration AlphaScreen™ kinase assay and a biophysical thermal shift assay, followed by X-ray crystallography, identified a number of chemically different ligand-efficient CHK2 hinge-binding scaffolds that have not been exploited in known CHK2 inhibitors. In addition, it showed that the use of these orthogonal techniques allowed efficient discrimination between genuine hit matter and false positives from each individual assay technology. Furthermore, the CHK2 crystal structures with a quinoxaline-based fragment and its follow-up compound highlight a hydrophobic area above the hinge region not previously explored in rational CHK2 inhibitor design, but which might be exploited to enhance both potency and selectivity of CHK2 inhibitors.


Asunto(s)
Adenosina Trifosfato/metabolismo , Quinasa de Punto de Control 2/química , Quinasa de Punto de Control 2/metabolismo , Modelos Moleculares , Conformación Proteica , Sitios de Unión/genética , Quinasa de Punto de Control 2/antagonistas & inhibidores , Cristalografía , Cristalografía por Rayos X , Estructura Molecular , Inhibidores de Proteínas Quinasas/química
5.
J Med Chem ; 55(22): 10229-40, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23082860

RESUMEN

Inhibitors of checkpoint kinase 1 (CHK1) are of current interest as potential antitumor agents, but the most advanced inhibitor series reported to date are not orally bioavailable. A novel series of potent and orally bioavailable 3-alkoxyamino-5-(pyridin-2-ylamino)pyrazine-2-carbonitrile CHK1 inhibitors was generated by hybridization of two lead scaffolds derived from fragment-based drug design and optimized for CHK1 potency and high selectivity using a cell-based assay cascade. Efficient in vivo pharmacokinetic assessment was used to identify compounds with prolonged exposure following oral dosing. The optimized compound (CCT244747) was a potent and highly selective CHK1 inhibitor, which modulated the DNA damage response pathway in human tumor xenografts and showed antitumor activity in combination with genotoxic chemotherapies and as a single agent.


Asunto(s)
Aminopiridinas/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química , Pirimidinas/farmacología , Administración Oral , Aminopiridinas/síntesis química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Niño , Neoplasias del Colon/enzimología , Daño del ADN/efectos de los fármacos , Diseño de Fármacos , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/enzimología , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/metabolismo , Pirimidinas/síntesis química , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Clin Cancer Res ; 18(20): 5650-61, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22929806

RESUMEN

PURPOSE: Many tumors exhibit defective cell-cycle checkpoint control and increased replicative stress. CHK1 is critically involved in the DNA damage response and maintenance of replication fork stability. We have therefore discovered a novel potent, highly selective, orally active ATP-competitive CHK1 inhibitor, CCT244747, and present its preclinical pharmacology and therapeutic activity. EXPERIMENTAL DESIGN: Cellular CHK1 activity was assessed using an ELISA assay, and cytotoxicity a SRB assay. Biomarker modulation was measured using immunoblotting, and cell-cycle effects by flow cytometry analysis. Single-agent oral CCT244747 antitumor activity was evaluated in a MYCN-driven transgenic mouse model of neuroblastoma by MRI and in genotoxic combinations in human tumor xenografts by growth delay. RESULTS: CCT244747 inhibited cellular CHK1 activity (IC(50) 29-170 nmol/L), significantly enhanced the cytotoxicity of several anticancer drugs, and abrogated drug-induced S and G(2) arrest in multiple tumor cell lines. Biomarkers of CHK1 (pS296 CHK1) activity and cell-cycle inactivity (pY15 CDK1) were induced by genotoxics and inhibited by CCT244747 both in vitro and in vivo, producing enhanced DNA damage and apoptosis. Active tumor concentrations of CCT244747 were obtained following oral administration. The antitumor activity of both gemcitabine and irinotecan were significantly enhanced by CCT244747 in several human tumor xenografts, giving concomitant biomarker modulation indicative of CHK1 inhibition. CCT244747 also showed marked antitumor activity as a single agent in a MYCN-driven neuroblastoma. CONCLUSION: CCT244747 represents the first structural disclosure of a highly selective, orally active CHK1 inhibitor and warrants further evaluation alone or combined with genotoxic anticancer therapies.


Asunto(s)
Aminopiridinas/administración & dosificación , Neoplasias Experimentales , Neuroblastoma , Proteínas Quinasas , Pirimidinas/administración & dosificación , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo
7.
J Med Chem ; 55(7): 3228-41, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22404346
8.
PLoS One ; 7(1): e28568, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253692

RESUMEN

Human cancers often contain genetic alterations that disable G1/S checkpoint control and loss of this checkpoint is thought to critically contribute to cancer generation by permitting inappropriate proliferation and distorting fate-driven cell cycle exit. The identification of cell permeable small molecules that activate the G1/S checkpoint may therefore represent a broadly applicable and clinically effective strategy for the treatment of cancer. Here we describe the identification of several novel small molecules that trigger G1/S checkpoint activation and characterise the mechanism of action for one, CCT020312, in detail. Transcriptional profiling by cDNA microarray combined with reverse genetics revealed phosphorylation of the eukaryotic initiation factor 2-alpha (EIF2A) through the eukaryotic translation initiation factor 2-alpha kinase 3 (EIF2AK3/PERK) as the mechanism of action of this compound. While EIF2AK3/PERK activation classically follows endoplasmic reticulum (ER) stress signalling that sets off a range of different cellular responses, CCT020312 does not trigger these other cellular responses but instead selectively elicits EIF2AK3/PERK signalling. Phosphorylation of EIF2A by EIF2A kinases is a known means to block protein translation and hence restriction point transit in G1, but further supports apoptosis in specific contexts. Significantly, EIF2AK3/PERK signalling has previously been linked to the resistance of cancer cells to multiple anticancer chemotherapeutic agents, including drugs that target the ubiquitin/proteasome pathway and taxanes. Consistent with such findings CCT020312 sensitizes cancer cells with defective taxane-induced EIF2A phosphorylation to paclitaxel treatment. Our work therefore identifies CCT020312 as a novel small molecule chemical tool for the selective activation of EIF2A-mediated translation control with utility for proof-of-concept applications in EIF2A-centered therapeutic approaches, and as a chemical starting point for pathway selective agent development. We demonstrate that consistent with its mode of action CCT020312 is capable of delivering potent, and EIF2AK3 selective, proliferation control and can act as a sensitizer to chemotherapy-associated stresses as elicited by taxanes.


Asunto(s)
Activadores de Enzimas/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo , Animales , Análisis por Conglomerados , Ciclina D1/metabolismo , ADN Complementario/genética , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Estrés del Retículo Endoplásmico/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/química , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Paclitaxel/farmacología , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
9.
J Med Chem ; 54(24): 8328-42, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-22111927

RESUMEN

Pyrazolopyridine inhibitors with low micromolar potency for CHK1 and good selectivity against CHK2 were previously identified by fragment-based screening. The optimization of the pyrazolopyridines to a series of potent and CHK1-selective isoquinolines demonstrates how fragment-growing and scaffold morphing strategies arising from a structure-based understanding of CHK1 inhibitor binding can be combined to successfully progress fragment-derived hit matter to compounds with activity in vivo. The challenges of improving CHK1 potency and selectivity, addressing synthetic tractability, and achieving novelty in the crowded kinase inhibitor chemical space were tackled by multiple scaffold morphing steps, which progressed through tricyclic pyrimido[2,3-b]azaindoles to N-(pyrazin-2-yl)pyrimidin-4-amines and ultimately to imidazo[4,5-c]pyridines and isoquinolines. A potent and highly selective isoquinoline CHK1 inhibitor (SAR-020106) was identified, which potentiated the efficacies of irinotecan and gemcitabine in SW620 human colon carcinoma xenografts in nude mice.


Asunto(s)
Antineoplásicos/síntesis química , Isoquinolinas/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/metabolismo , Pirazinas/síntesis química , Piridinas/síntesis química , Adenosina Trifosfato/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Disponibilidad Biológica , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Isoquinolinas/química , Isoquinolinas/farmacología , Ratones , Ratones Desnudos , Conformación Molecular , Trasplante de Neoplasias , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/química , Pirazinas/farmacología , Piridinas/química , Piridinas/farmacología , Estereoisomerismo , Relación Estructura-Actividad , Trasplante Heterólogo
10.
J Med Chem ; 54(6): 1626-39, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21366329

RESUMEN

We describe herein the structure-activity relationship (SAR) and cocrystal structures of a series of Nek2 inhibitors derived from the published polo-like kinase 1 (Plk1) inhibitor (R)-1. Our studies reveal a nonlinear SAR for Nek2 and our cocrystal structures show that compounds in this series bind to a DFG-out conformation of Nek2 without extending into the enlarged back pocket commonly found in this conformation. These observations were further investigated, and structure-based design led to Nek2 inhibitors derived from (R)-1 with more than a hundred-fold selectivity against Plk1.


Asunto(s)
Benzamidas/síntesis química , Bencimidazoles/síntesis química , Modelos Moleculares , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Benzamidas/química , Benzamidas/farmacología , Bencimidazoles/química , Bencimidazoles/farmacología , Sitios de Unión , Cristalografía por Rayos X , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Quinasas Relacionadas con NIMA , Fosforilación , Unión Proteica , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Estereoisomerismo , Relación Estructura-Actividad
11.
J Med Chem ; 54(2): 580-90, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21186793

RESUMEN

Structure-based design was applied to the optimization of a series of 2-(quinazolin-2-yl)phenols to generate potent and selective ATP-competitive inhibitors of the DNA damage response signaling enzyme checkpoint kinase 2 (CHK2). Structure-activity relationships for multiple substituent positions were optimized separately and in combination leading to the 2-(quinazolin-2-yl)phenol 46 (IC(50) 3 nM) with good selectivity for CHK2 against CHK1 and a wider panel of kinases and with promising in vitro ADMET properties. Off-target activity at hERG ion channels shown by the core scaffold was successfully reduced by the addition of peripheral polar substitution. In addition to showing mechanistic inhibition of CHK2 in HT29 human colon cancer cells, a concentration dependent radioprotective effect in mouse thymocytes was demonstrated for the potent inhibitor 46 (CCT241533).


Asunto(s)
Fenoles/síntesis química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinazolinas/síntesis química , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular , Quinasa de Punto de Control 2 , Cristalografía por Rayos X , Diseño de Fármacos , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/metabolismo , Células HT29 , Humanos , Técnicas In Vitro , Ratones , Modelos Moleculares , Fenoles/química , Fenoles/farmacología , Unión Proteica , Quinazolinas/química , Quinazolinas/farmacología , Protectores contra Radiación/síntesis química , Protectores contra Radiación/química , Protectores contra Radiación/farmacología , Estereoisomerismo , Relación Estructura-Actividad , Timo/citología , Timo/efectos de los fármacos , Timo/efectos de la radiación
12.
J Med Chem ; 53(21): 7682-98, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20936789

RESUMEN

We report herein the first systematic exploration of inhibitors of the mitotic kinase Nek2. Starting from HTS hit aminopyrazine 2, compounds with improved activity were identified using structure-based design. Our structural biology investigations reveal two notable observations. First, 2 and related compounds bind to an unusual, inactive conformation of the kinase which to the best of our knowledge has not been reported for other types of kinase inhibitors. Second, a phenylalanine residue at the center of the ATP pocket strongly affects the ability of the inhibitor to bind to the protein. The implications of these observations are discussed, and the work described here defines key features for potent and selective Nek2 inhibition, which will aid the identification of more advanced inhibitors of Nek2.


Asunto(s)
Modelos Moleculares , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazinas/síntesis química , Cristalografía por Rayos X , Humanos , Quinasas Relacionadas con NIMA , Fosforilación , Unión Proteica , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Pirazinas/química , Estereoisomerismo , Relación Estructura-Actividad
13.
Bioorg Med Chem Lett ; 20(14): 4045-9, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20561787

RESUMEN

A range of 3,6-di(hetero)arylimidazo[1,2-a]pyrazine ATP-competitive inhibitors of CHK1 were developed by scaffold hopping from a weakly active screening hit. Efficient synthetic routes for parallel synthesis were developed to prepare analogues with improved potency and ligand efficiency against CHK1. Kinase profiling showed that the imidazo[1,2-a]pyrazines could inhibit other kinases, including CHK2 and ABL, with equivalent or better potency depending on the pendant substitution. These 3,6-di(hetero)aryl imidazo[1,2-a]pyrazines appear to represent a general kinase inhibitor scaffold.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/química , Pirazinas/farmacología , Cristalografía por Rayos X , Diseño de Fármacos , Evaluación Preclínica de Medicamentos
14.
J Med Chem ; 53(5): 2239-49, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20151677

RESUMEN

Protein kinase B (PKB or Akt) is an important component of intracellular signaling pathways regulating growth and survival. Signaling through PKB is frequently deregulated in cancer, and inhibitors of PKB therefore have potential as antitumor agents. The optimization of lipophilic substitution within a series of 4-benzyl-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidin-4-amines provided ATP-competitive, nanomolar inhibitors with up to 150-fold selectivity for inhibition of PKB over the closely related kinase PKA. Although active in cellular assays, compounds containing 4-amino-4-benzylpiperidines underwent metabolism in vivo, leading to rapid clearance and low oral bioavailability. Variation of the linker group between the piperidine and the lipophilic substituent identified 4-amino-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamides as potent and orally bioavailable inhibitors of PKB. Representative compounds modulated biomarkers of signaling through PKB in vivo and strongly inhibited the growth of human tumor xenografts in nude mice at well-tolerated doses.


Asunto(s)
Antineoplásicos/síntesis química , Piperidinas/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/síntesis química , Pirroles/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Semivida , Humanos , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Ratones , Ratones Desnudos , Piperidinas/química , Piperidinas/farmacocinética , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirimidinas/química , Pirimidinas/farmacocinética , Pirroles/química , Pirroles/farmacocinética , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Cancer Ther ; 9(1): 89-100, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20053762

RESUMEN

Genotoxic antitumor agents continue to be the mainstay of current cancer chemotherapy. These drugs cause DNA damage and activate numerous cell cycle checkpoints facilitating DNA repair and the maintenance of genomic integrity. Most human tumors lack functional p53 and consequently have compromised G(1)-S checkpoint control. This has led to the hypothesis that S and G(2)-M checkpoint abrogation may selectively enhance genotoxic cell killing in a p53-deficient background, as normal cells would be rescued at the G(1)-S checkpoint. CHK1 is a serine/threonine kinase associated with DNA damage-linked S and G(2)-M checkpoint control. SAR-020106 is an ATP-competitive, potent, and selective CHK1 inhibitor with an IC(50) of 13.3 nmol/L on the isolated human enzyme. This compound abrogates an etoposide-induced G(2) arrest with an IC(50) of 55 nmol/L in HT29 cells, and significantly enhances the cell killing of gemcitabine and SN38 by 3.0- to 29-fold in several colon tumor lines in vitro and in a p53-dependent fashion. Biomarker studies have shown that SAR-020106 inhibits cytotoxic drug-induced autophosphorylation of CHK1 at S296 and blocks the phosphorylation of CDK1 at Y15 in a dose-dependent fashion both in vitro and in vivo. Cytotoxic drug combinations were associated with increased gammaH2AX and poly ADP ribose polymerase cleavage consistent with the SAR-020106-enhanced DNA damage and tumor cell death. Irinotecan and gemcitabine antitumor activity was enhanced by SAR-020106 in vivo with minimal toxicity. SAR-020106 represents a novel class of CHK1 inhibitors that can enhance antitumor activity with selected anticancer drugs in vivo and may therefore have clinical utility.


Asunto(s)
Isoquinolinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Pirazinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/metabolismo , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Camptotecina/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sinergismo Farmacológico , Fase G2/efectos de los fármacos , Humanos , Irinotecán , Isoquinolinas/administración & dosificación , Isoquinolinas/química , Isoquinolinas/farmacocinética , Ratones , Ratones Desnudos , Mutágenos/toxicidad , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazinas/administración & dosificación , Pirazinas/química , Pirazinas/farmacocinética , Gemcitabina
16.
J Med Chem ; 52(15): 4810-9, 2009 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-19572549

RESUMEN

Checkpoint kinase 1 (CHK1) is an oncology target of significant current interest. Inhibition of CHK1 abrogates DNA damage-induced cell cycle checkpoints and sensitizes p53 deficient cancer cells to genotoxic therapies. Using template screening, a fragment-based approach to small molecule hit generation, we have identified multiple CHK1 inhibitor scaffolds suitable for further optimization. The sequential combination of in silico low molecular weight template selection, a high concentration biochemical assay and hit validation through protein-ligand X-ray crystallography provided 13 template hits from an initial in silico screening library of ca. 15000 compounds. The use of appropriate counter-screening to rule out nonspecific aggregation by test compounds was essential for optimum performance of the high concentration bioassay. One low molecular weight, weakly active purine template hit was progressed by iterative structure-based design to give submicromolar pyrazolopyridines with good ligand efficiency and appropriate CHK1-mediated cellular activity in HT29 colon cancer cells.


Asunto(s)
Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Evaluación Preclínica de Medicamentos , Células HT29 , Humanos , Enlace de Hidrógeno , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química , Relación Estructura-Actividad
17.
Bioorg Med Chem ; 17(2): 467-74, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19101154

RESUMEN

High-throughput screening led to the identification of isothiazolones 1 and 2 as inhibitors of histone acetyltransferase (HAT) with IC50s of 3 microM and 5 microM, respectively. Analogues of these hit compounds with variations of the N-phenyl group, and with variety of substituents at C-4, C-5 of the thiazolone ring, were prepared and assayed for inhibition of the HAT enzyme PCAF. Potency is modestly favoured when the N-aryl group is electron deficient (4-pyridyl derivative 10 has IC(50)=1.5 microM); alkyl substitution at C-4 has little effect, whilst similar substitution at C-5 causes a significant drop in potency. The ring-fused compound 38 has activity (IC(50)=6.1 microM) to encourage further exploration of this bicyclic structure. The foregoing SAR is consistent with an inhibitory mechanism involving cleavage of the S-N bond of the isothiazolone ring by a catalytically important thiol residue.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Histona Acetiltransferasas/antagonistas & inhibidores , Tiazoles/síntesis química , Animales , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Relación Estructura-Actividad , Tiazoles/farmacología , Factores de Transcripción p300-CBP/antagonistas & inhibidores
18.
J Med Chem ; 51(7): 2147-57, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18345609

RESUMEN

Fragment-based screening identified 7-azaindole as a protein kinase B inhibitor scaffold. Fragment elaboration using iterative crystallography of inhibitor-PKA-PKB chimera complexes efficiently guided improvements in the potency and selectivity of the compounds, resulting in the identification of nanomolar 6-(piperidin-1-yl)purine, 4-(piperidin-1-yl)-7-azaindole, and 4-(piperidin-1-yl)pyrrolo[2,3- d]pyrimidine inhibitors of PKBbeta with antiproliferative activity and showing pathway inhibition in cells. A divergence in the binding mode was seen between 4-aminomethylpiperidine and 4-aminopiperidine containing molecules. Selectivity for PKB vs PKA was observed with 4-aminopiperidine derivatives, and the most PKB-selective inhibitor (30-fold) showed significantly different bound conformations between PKA and PKA-PKB chimera.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirimidinas/química , Pirimidinas/farmacología , Pirroles/química , Pirroles/farmacología , Animales , Sitios de Unión , Línea Celular Tumoral , Cromatografía Liquida/métodos , Cristalografía por Rayos X , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Humanos , Ligandos , Espectroscopía de Resonancia Magnética/métodos , Espectrometría de Masas/métodos , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Estructura Molecular , Pirimidinas/metabolismo , Pirroles/metabolismo , Estereoisomerismo , Relación Estructura-Actividad , Especificidad por Sustrato
19.
J Med Chem ; 50(10): 2289-92, 2007 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-17451235
20.
EMBO J ; 25(13): 3179-90, 2006 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-16794575

RESUMEN

The protein kinase Chk2 (checkpoint kinase 2) is a major effector of the replication checkpoint. Chk2 activation is initiated by phosphorylation of Thr68, in the serine-glutamine/threonine-glutamine cluster domain (SCD), by ATM. The phosphorylated SCD-segment binds to the FHA domain of a second Chk2 molecule, promoting dimerisation of the protein and triggering phosphorylation of the activation segment/T-loop in the kinase domain. We have now determined the structure of the kinase domain of human Chk2 in complexes with ADP and a small-molecule inhibitor debromohymenialdisine. The structure reveals a remarkable dimeric arrangement in which T-loops are exchanged between protomers, to form an active kinase conformation in trans. Biochemical data suggest that this dimer is the biologically active state promoted by ATM-phosphorylation, and also suggests a mechanism for dimerisation-driven activation of Chk2 by trans-phosphorylation.


Asunto(s)
Daño del ADN , Modelos Moleculares , Proteínas Serina-Treonina Quinasas/química , Transactivadores , Adenosina Difosfato/química , Proteínas de la Ataxia Telangiectasia Mutada , Azepinas/química , Dominio Catalítico , Proteínas de Ciclo Celular/química , Quinasa de Punto de Control 2 , Proteínas de Unión al ADN/química , Dimerización , Activación Enzimática , Humanos , Fosforilación , Conformación Proteica , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Pirroles/química , Transducción de Señal , Proteínas Supresoras de Tumor/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...