Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nephrology (Carlton) ; 16(4): 410-6, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21518118

RESUMO

AIM: The cyclin-dependent kinase inhibitor, seliciclib (R-roscovitine, CYC202), has anti-proliferative activity through its inhibition of cyclin-dependent kinase 2. We hypothesized that treatment with seliciclib would reduce glomerular macrophage numbers and glomerular crescent formation in experimental crescentic glomerulonephritis even when treatment is started after onset of disease. METHOD: Nephrotoxic nephritis (NTN) was induced in Wistar Kyoto rats. In experiment 1, seliciclib (150 mg/kg per day) was given by oral gavage from 1 h before induction of NTN and continued to day 14. In experiment 2, treatment was started on day 4 of NTN and continued to day 14 in order to examine the effect of seliciclib in established glomerulonephritis. RESULTS: In experiment 1, seliciclib reduced proteinuria (119.5 ± 13.9 vs 191.4 ± 18.8 mg/day, P < 0.01), serum creatinine (54.0 ± 3.0 vs 81.0 ± 2.5 µmol/L, P < 0.005) and glomerular crescent score (23.9 ± 2.1 vs 44.6 ± 2.2, P < 0.005) in comparison with controls. In experiment 2, seliciclib ameliorated established glomerulonephritis, with reduction in proteinuria (58 ± 16 vs 165 ± 13 mg/day, P < 0.005), serum creatinine (39 ± 3 vs 62 ± 5 µmol/L, P < 0.05), glomerular macrophage numbers (6.8 ± 2.5 vs 18.5 ± 1.2 ED1+ cells per glomerular cross section, P < 0.05), glomerular cell proliferation (1.2 ± 0.37 vs 4.2 ± 0.80 bromodeoxyuridine (BrdU)+ cells per glomerular section, P < 0.05) and crescent score (10.8 ± 1.6 vs 43.9 ± 1.4, P < 0.05), in comparison with the controls. CONCLUSION: Seliciclib is effective in both prevention and treatment of established crescentic glomerulonephritis in Wistar Kyoto rats, in association with a reduction in the number of glomerular macrophages. We suggest that seliciclib, or other cyclin-dependent kinase inhibitors, may represent a novel therapeutic approach for patients with proliferative glomerulonephritis.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Glomerulonefrite/tratamento farmacológico , Glomérulos Renais/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Administração Oral , Animais , Proliferação de Células/efeitos dos fármacos , Creatinina/sangue , Quinases Ciclina-Dependentes/metabolismo , Modelos Animais de Doenças , Glomerulonefrite/enzimologia , Glomerulonefrite/patologia , Glomerulonefrite/prevenção & controle , Glomérulos Renais/enzimologia , Glomérulos Renais/patologia , Macrófagos/patologia , Masculino , Inibidores de Proteínas Quinases/administração & dosagem , Proteinúria/tratamento farmacológico , Proteinúria/enzimologia , Purinas/administração & dosagem , Ratos , Ratos Endogâmicos WKY , Roscovitina , Índice de Gravidade de Doença , Fatores de Tempo
2.
Clin Cancer Res ; 14(13): 4326-35, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18594016

RESUMO

PURPOSE: The aims of this study were to investigate whether the cyclin-dependent kinase inhibitor seliciclib could synergize with agents that target ErbB receptors and to elucidate the molecular mechanism of the observed synergy. EXPERIMENTAL DESIGN: Synergy between seliciclib and ErbB receptor targeted agents was investigated in various cell lines using the Calcusyn median effect model. The molecular mechanism of the observed synergy was studied in cultured cells, and the combination of seliciclib and the epidermal growth factor receptor (EGFR) inhibitor erlotinib was evaluated in an H358 xenograft model. RESULTS: Seliciclib synergized with the anti-HER2 antibody trastuzumab in a breast cancer cell line, which overexpresses the HER2 receptor, and with the erlotinib analogue AG1478 in non-small cell lung cancer cell lines. In the H358 non-small cell lung cancer cell line, synergy involved decreased signaling from the EGFR, with AG1478 directly inhibiting kinase activity while seliciclib decreased the levels of key components of the receptor signaling pathway, resulting in enhanced loss of phosphorylated extracellular signal-regulated kinase and cyclin D1. The combination of seliciclib and erlotinib was evaluated further in an H358 xenograft and shown to be significantly more active than either agent alone. An enhanced loss of cyclin D1 was also seen in vivo. CONCLUSIONS: This is the first report that investigates combining seliciclib with an EGFR inhibitor. The combination decreased signaling from the EGFR in vitro and in vivo and was effective in cell lines containing either wild-type or mutant EGFR, suggesting that it may expand the range of tumors that respond to erlotinib, and therefore, such combinations are worth exploring in the clinic.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Purinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Concentração Inibidora 50 , Camundongos , Modelos Biológicos , Mutação , Transplante de Neoplasias , Receptor ErbB-2/metabolismo , Roscovitina
3.
Drug Metab Dispos ; 36(3): 561-70, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18048486

RESUMO

Seliciclib (R-roscovitine, CYC202) is a small molecule inhibitor of cyclin-dependent kinases currently in phase II clinical trials as an anticancer agent. We examined the metabolism of seliciclib in vitro and in vivo. Using radiolabeled seliciclib we found that cytochrome P450 (P450)-mediated metabolism in liver microsomes from human, rat, mouse, rabbit, monkey, and dog was rapid to a number of metabolic species, one of the most prevalent being a carboxylate previously identified in urine from rats and mice dosed with seliciclib. Metabolism was fastest in mouse microsomes and slowest in microsomes from dog. Using characterized human microsomes, we identified the P450s responsible for this metabolism as CYP3A4 and CYP2B6. Glucuronidation of seliciclib and its metabolites was shown to be a major elimination process in bile duct-cannulated rats dosed with [(14)C]seliciclib at 10 mg/kg. Elimination by the fecal route accounted for up to 65% of the administered dose, whereas urinary excretion accounted for up to 43%. Almost half of the administered dose was found to be eliminated via the bile, and elimination was found to be rapid, with up to 88% of the dose being excreted within the first 24 h. Preliminary experiments indicated that UDP-glucuronosyltransferase (UGT) 1A3, 1A9, and 2B7 were involved in the conjugation of seliciclib. Seliciclib was further shown in vitro to inhibit the activity of some of the enzymes responsible for its metabolism. Cytochrome P450s CYP3A4 and CYP2C9 and UGT1A1 were all inhibited at concentrations achieved in human trials, which raises the possibility of drug-drug interactions in the clinic.


Assuntos
Inibidores de Proteínas Quinases/metabolismo , Purinas/metabolismo , Animais , Bile/metabolismo , Radioisótopos de Carbono , Cromatografia Líquida de Alta Pressão , Quinases Ciclina-Dependentes/antagonistas & inibidores , Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Fezes , Feminino , Glucuronídeos/metabolismo , Humanos , Macaca fascicularis , Masculino , Camundongos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/urina , Purinas/farmacocinética , Purinas/urina , Coelhos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Roscovitina , Especificidade da Espécie
4.
Clin Cancer Res ; 11(13): 4875-87, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16000586

RESUMO

PURPOSE: To investigate pharmacokinetic-pharmacodynamic relationships for the trisubstituted aminopurine cyclin-dependent kinase inhibitors olomoucine, bohemine, and CYC202 (R-roscovitine; seliciclib) in the HCT116 human colon carcinoma model. EXPERIMENTAL DESIGN: The in vitro activity of the agents was determined in a human tumor panel using the sulforhodamine B assay. The concentration and time dependence was established in HCT116 cells. Molecular biomarkers, including RB phosphorylation and cyclin expression, were assessed by Western blotting. Pharmacokinetic properties were characterized in mice following analysis by liquid chromatography-tandem mass spectrometry. Based on these studies, a dosing regimen was developed for CYC202 that allowed therapeutic exposures in the HCT116 tumor xenograft. RESULTS: The antitumor potency of the agents in vitro was in the order olomoucine (IC50, 56 micromol/L) < bohemine (IC50, 27 micromol/L) < CYC202 (IC50, 15 micromol/L), corresponding to their activities as cyclin-dependent kinase inhibitors. Antitumor activity increased with exposure time up to 16 hours. The agents caused inhibition of RB and RNA polymerase II phosphorylation and depletion of cyclins. They exhibited relatively rapid clearance following administration to mice. CYC202 displayed the slowest clearance from plasma and the highest tumor uptake, with oral bioavailability of 86%. Oral dosing of CYC202 gave active concentrations in the tumor, modulation of pharmacodynamic markers, and inhibition of tumor growth. CONCLUSIONS: CYC202 showed therapeutic activity on human cancer cell lines in vitro and on xenografts. Pharmacodynamic markers are altered in vitro and in vivo, consistent with the inhibition of cyclin-dependent kinases. Such markers may be potentially useful in the clinical development of CYC202 and other cyclin-dependent kinase inhibitors.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/farmacocinética , Neoplasias Experimentais/metabolismo , Animais , Área Sob a Curva , Western Blotting , Proteína Quinase CDC2/antagonistas & inibidores , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Feminino , Células HCT116 , Humanos , Concentração Inibidora 50 , Cinetina/farmacocinética , Cinetina/farmacologia , Dose Máxima Tolerável , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/patologia , Fosforilação/efeitos dos fármacos , Purinas/farmacocinética , Purinas/farmacologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Proteína do Retinoblastoma/metabolismo , Roscovitina , Fatores de Tempo , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Structure ; 11(4): 399-410, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12679018

RESUMO

A family of 4-heteroaryl-2-amino-pyrimidine CDK2 inhibitor lead compounds was discovered with the new database-mining program LIDAEUS through in silico screening. Four compounds with IC(50) values ranging from 17 to 0.9 microM were selected for X-ray crystal analysis. Two distinct binding modes are observed, one of which resembles the hydrogen bonding pattern of bound ATP. In the second binding mode, the ligands trigger a conformational change in the activation T loop by inducing movement of Lys(33) and Asp(145) side chains. The family of molecules discovered provides an excellent starting point for the design and synthesis of tight binding inhibitors, which may lead to a new class of antiproliferative drugs.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Estrutura Secundária de Proteína , Software , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Quinases Ciclina-Dependentes/química , Quinases Ciclina-Dependentes/metabolismo , Bases de Dados de Proteínas , Humanos , Ligação de Hidrogênio , Ligantes , Substâncias Macromoleculares , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Piridinas/química , Piridinas/metabolismo
6.
Mol Cancer Ther ; 4(1): 125-39, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15657360

RESUMO

R-roscovitine (seliciclib, CYC202) is a cyclin-dependent kinase inhibitor currently in phase II clinical trials in patients with cancer. Here, we describe its mouse metabolism and pharmacokinetics as well as the identification of the principal metabolites in hepatic microsomes, plasma, and urine. Following microsomal incubation of R-roscovitine at 10 microg/mL (28 micromol/L) for 60 minutes, 86.7% of the parent drug was metabolized and 60% of this loss was due to formation of one particular metabolite. This was identified as the carboxylic acid resulting from oxidation of the hydroxymethyl group of the amino alcohol substituent at C2 of the purine core present in R-roscovitine. Identification was confirmed by chemical synthesis and comparison of an authentic sample of the R-roscovitine-derived carboxylate metabolite (COOH-R-roscovitine). Other minor metabolites were identified as C8-oxo-R-roscovitine and N9-desisopropyl-R-roscovitine; these accounted for 4.9% and 2.6% of the parent, respectively. The same metabolic pattern was observed in vivo, with a 4.5-fold lower AUC(infinity) for R-roscovitine (38 micromol/L/h) than for COOH-R-roscovitine (174 micromol/L/h). Excretion of R-roscovitine in the urine up to 24 hours post-dosing accounted for an average of only 0.02% of the administered dose of 50 mg/kg, whereas COOH-R-roscovitine represented 65% to 68% of the dose irrespective of the route of administration (i.v., i.p., or p.o.). A partially deuterated derivative (R-roscovitine-d9) was synthesized to investigate if formation of COOH-R-roscovitine could be inhibited by replacement of metabolically labile protons with deuterium. After 60 minutes of incubation of R-roscovitine-d9 or R-roscovitine with mouse liver microsomes, formation of COOH-R-roscovitine-d9 was decreased by approximately 24% compared with the production of COOH-R-roscovitine. In addition, the levels of R-roscovitine-d9 remaining were 33% higher than those of R-roscovitine. However, formation of several minor R-roscovitine metabolites was enhanced with R-roscovitine-d9, suggesting that metabolic switching from the major carbinol oxidation pathway had occurred. Synthetic COOH-R-roscovitine and C8-oxo-R-roscovitine were tested in functional cyclin-dependent kinase assays and shown to be less active than R-roscovitine, confirming that these metabolic reactions are deactivation pathways.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Purinas/farmacocinética , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Biotransformação , Espectrometria de Massas , Camundongos , Microssomos Hepáticos/metabolismo , Purinas/química , Roscovitina , Distribuição Tecidual
7.
Chem Biol ; 17(10): 1111-21, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-21035734

RESUMO

The main difficulty in the development of ATP antagonist kinase inhibitors is target specificity, since the ATP-binding motif is present in many proteins. We introduce a strategy that has allowed us to identify compounds from a kinase inhibitor library that block the cyclin-dependent kinases responsible for regulating transcription, i.e., CDK7 and especially CDK9. The screening cascade employs cellular phenotypic assays based on mitotic index and nuclear p53 protein accumulation. This permitted us to classify compounds into transcriptional, cell cycle, and mitotic inhibitor groups. We describe the characterization of the transcriptional inhibitor class in terms of kinase inhibition profile, cellular mode of action, and selectivity for transformed cells. A structural selectivity rationale was used to optimize potency and biopharmaceutical properties and led to the development of a transcriptional inhibitor, 3,4-dimethyl-5-[2-(4-piperazin-1-yl-phenylamino)-pyrimidin-4-yl]-3H-thiazol-2-one, with anticancer activity in animal models.


Assuntos
Antineoplásicos/química , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Pirimidinas/química , Animais , Antineoplásicos/uso terapêutico , Apoptose , Sítios de Ligação , Linhagem Celular Tumoral , Simulação por Computador , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Humanos , Leucemia/tratamento farmacológico , Camundongos , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
8.
J Med Chem ; 53(11): 4367-78, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20462263

RESUMO

Through cell-based screening of our kinase-directed compound collection, we discovered that a subset of N-phenyl-4-(thiazol-5-yl)pyrimidin-2-amines were potent cytotoxic agents against cancer cell lines, suppressed mitotic histone H3 phosphorylation, and caused aberrant mitotic phenotypes. It was subsequently established that these compounds were in fact potent inhibitors of aurora A and B kinases. It was shown that potency and selectivity of aurora kinase inhibition correlated with the presence of a substituent at the aniline para-position in these compounds. The anticancer effects of lead compound 4-methyl-5-(2-(4-morpholinophenylamino)pyrimidin-4-yl)thiazol-2-amine (18; K(i) values of 8.0 and 9.2 nM for aurora A and B, respectively) were shown to emanate from cell death following mitotic failure and increased polyploidy as a consequence of cellular inhibition of aurora A and B kinases. Preliminary in vivo assessment showed that compound 18 was orally bioavailable and possessed anticancer activity. Compound 18 (CYC116) is currently undergoing phase I clinical evaluation in cancer patients.


Assuntos
Descoberta de Drogas/métodos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Tiazóis/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Aurora Quinase A , Aurora Quinases , Ligação Competitiva , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Camundongos , Mitose/efeitos dos fármacos , Modelos Moleculares , Conformação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/química , Pirimidinas/metabolismo , Pirimidinas/farmacocinética , Especificidade por Substrato , Tiazóis/química , Tiazóis/metabolismo , Tiazóis/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Expert Opin Drug Discov ; 3(1): 131-43, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23480144

RESUMO

Seliciclib is an inhibitor of cyclin-dependent kinases 2, 7 and 9. Its primary mechanism of action is the inhibition of transcription, resulting in the selective downregulation of rapidly cycling mRNA transcripts, including Mcl-1 and cyclin D1. It possesses antitumour activity as a single agent and also synergises with a wide range of cytotoxic and targeted drugs. Seliciclib has high oral bioavailability and is in clinical development in a capsule formulation. The clinical dose has been determined in Phase I clinical trials for schedules of 3 - 10 consecutive days per cycle of 2 or 3 weeks duration. Its major clinical toxicities include nausea, vomiting, asthenia, hypokalaemia, elevation of creatinine levels and liver function tests, which are reversible after cessation of dosing. Seliciclib is non-myelosuppressive and does not cause intestinal toxicity. Phase II trials have commenced in non-small cell lung cancer and will be initiated shortly in nasopharyngeal carcinoma.

10.
Int J Cancer ; 102(5): 463-8, 2002 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-12432547

RESUMO

CDK2 inhibitors have been proposed as effective anti-cancer therapeutics. We show here that CYC202 (R-roscovitine) is a potent inhibitor of recombinant CDK2/cyclin E kinase activity (IC(50) = 0.10 microM) with an average cytotoxic IC(50) of 15.2 microM in a panel of 19 human tumour cell lines, and we also demonstrate selectivity for rapidly proliferating cells over non-proliferating cells. A study of the cell cycle effects of CYC202 in Lovo colorectal carcinoma cells showed that the major effect was not the predicted arrest in one part of the cycle, but rather an induction of cell death from all compartments of the cell cycle. The maximum tolerated dose given intravenously to mice was in excess of 20 mg/kg. Doses up to 2,000 mg/kg were tolerated when administered orally in mice. Following repeated intraperitoneal administration (3 times daily for 5 days) of 100 mg/kg to nude mice bearing the Lovo human colorectal tumour, CYC202 induced a significant antitumour effect with a 45% reduction in tumour growth compared to controls. A second experiment using the human uterine xenograft MESSA-DX5 treated with orally administered CYC202 (500 mg/kg 3 times daily for 4 days) also exhibited a significant reduction in the rate of growth of the tumour (62%). These data, showing enzyme and cellular potency together with antitumour activity, confirm the potential of CDK2 inhibitors such as CYC202 as anticancer drugs.


Assuntos
Antineoplásicos/uso terapêutico , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Neoplasias/tratamento farmacológico , Purinas/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Ciclina E , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteínas Recombinantes , Roscovitina , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA