Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Lett ; 381(1): 49-57, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27461583

RESUMO

With conventional anticancer agents for non-small cell lung cancer (NSCLC) reaching therapeutic ceiling, the novel combination using histone deacetylase inhibitor, PXD101 (Belinostat(®)), and CDK inhibitor, CYC202 (Seliciclib(®)), was investigated as an alternative anticancer strategy. At clinically achievable concentration of CYC202 (15 µM), combination therapy resulted in significant reduction in cell proliferation (IC50 = 3.67 ± 0.80 µM, p < 0.05) compared with PXD101 alone (IC50 = 6.56 ± 0.42 µM) in p53 wild-type A549 cells. Significant increase in apoptosis that occurred independently of cell cycle arrest was observed after concurrent treatment. This result was corroborated by greater formation of cleaved caspase-8, caspase-3 and PARP. Up-regulation of p53 and truncated BID protein levels was seen while Mcl-1 and XIAP protein levels were down-regulated upon combined treatment. Further analysis of apoptotic pathways revealed that caspase inhibitors, but not p53 silencing, significantly abrogated the cytotoxic enhancement. Moreover, the enhanced efficacy of this combination was additionally confirmed in p53 null H2444 cells, suggesting the potential of this combination for treatment of NSCLC that are not amenable to effects of conventional p53-inducing agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/agonistas , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Sulfonamidas/farmacologia , Células A549 , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Inibidores de Caspase/farmacologia , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Interferência de RNA , Roscovitina , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
2.
PLoS One ; 10(3): e0118553, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25793887

RESUMO

A novel, rapid and sensitive liquid chromatography-tandem mass spectrometric (LC-MS/MS) method was developed and validated for the evaluation of exemestane pharmacokinetics and its metabolites, 17ß-dihydroexemestane (active metabolite) and 17ß-dihydroexemestane-17-O-ß-D-glucuronide (inactive metabolite) in human plasma. Their respective D3 isotopes were used as internal standards. Chromatographic separation of analytes was achieved using Thermo Fisher BDS Hypersil C18 analytic HPLC column (100 × 2.1 mm, 5 µm). The mobile phase was delivered at a rate of 0.5 mL/min by gradient elution with 0.1% aqueous formic acid and acetonitrile. The column effluents were detected by API 4000 triple quadrupole mass spectrometer using electrospray ionisation (ESI) and monitored by multiple reaction monitoring (MRM) in positive mode. Mass transitions 297 > 121 m/z, 300 > 121 m/z, 299 > 135 m/z, 302 > 135 m/z, 475 > 281 m/z, and 478 > 284 m/z were monitored for exemestane, exemestane-d3, 17ß-dihydroexemestane, 17ß-dihydroexemestane-d3, 17ß-dihydroexemestane-17-O-ß-D-glucuronide, and 17ß-dihydroexemestane-17-O-ß-D-glucuronide-d3 respectively. The assay demonstrated linear ranges of 0.4-40.0 ng/mL, for exemestane; and 0.2-15.0 ng/mL, for 17ß-dihydroexemestane and 17ß-dihydroexemestane-17-O-ß-D-glucuronide, with coefficient of determination (r2) of > 0.998. The precision (coefficient of variation) were ≤10.7%, 7.7% and 9.5% and the accuracies ranged from 88.8 to 103.1% for exemestane, 98.5 to 106.1% for 17ß-dihydroexemestane and 92.0 to 103.2% for 17ß-dihydroexemestane-17-O-ß-D-glucuronide. The method was successfully applied to a pharmacokinetics/dynamics study in breast cancer patients receiving exemestane 25 mg daily orally. For a representative patient, 20.7% of exemestane in plasma was converted into 17ß-dihydroexemestane and 29.0% of 17ß-dihydroexemestane was inactivated as 17ß-dihydroexemestane-17-O-ß-D-glucuronide 24 hours after ingestion of exemestane, suggesting that altered 17-dihydroexemestane glucuronidation may play an important role in determining effect of exemestane against breast cancer cells.


Assuntos
Androstadienos/sangue , Androstadienos/farmacocinética , Cromatografia Líquida/métodos , Glucuronídeos/sangue , Glucuronídeos/farmacocinética , Espectrometria de Massas em Tandem/métodos , Congelamento , Humanos , Redes e Vias Metabólicas , Controle de Qualidade , Padrões de Referência , Reprodutibilidade dos Testes
3.
J Mass Spectrom ; 48(3): 406-12, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23494799

RESUMO

A sensitive analytical method has been developed and validated for the quantification of L-ergothioneine in human plasma and erythrocytes by liquid chromatography-tandem mass spectrometry. A commercially available isotope-labeled L-ergothioneine-d9 is used as the internal standard. A simple protein precipitation with acetonitrile is utilized for bio-sample preparation prior to analysis. Chromatographic separation of L-ergothioneine is conducted using gradient elution on Alltime C18 (150 mm × 2.1 mm, 5 µ). The run time is 6 min at a constant flow rate of 0.45 ml/min. The mass spectrometer is operated under a positive electrospray ionization condition with multiple reaction monitoring mode. The mass transitions of L-ergothioneine and L-ergothioneine-d9 are m/z 230 > 127 and m/z 239 > 127, respectively. Excellent linearity [coefficient of determination (r(2)) ≥ 0.9998] can be achieved for L-ergothioneine quantification at the ranges of 10 to 10,000 ng/ml, with the intra-day and inter-day precisions at 0.9-3.9% and 1.3-5.7%, respectively, and the accuracies for all quality control samples between 94.5 and 101.0%. This validated analytical method is suitable for pharmacokinetic monitoring of L-ergothioneine in human and erythrocytes. Based on the determination of bio-samples from five healthy subjects, the mean concentrations of L-ergothioneine in plasma and erythrocytes are 107.4 ± 20.5 ng/ml and 1285.0 ± 1363.0 ng/ml, respectively.


Assuntos
Antioxidantes/farmacocinética , Ergotioneína/farmacocinética , Espectrometria de Massas em Tandem/métodos , Antioxidantes/análise , Cromatografia Líquida de Alta Pressão/métodos , Ergotioneína/análise , Ergotioneína/sangue , Eritrócitos/química , Humanos , Modelos Lineares , Sensibilidade e Especificidade
4.
PLoS One ; 8(1): e54522, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23382909

RESUMO

UNLABELLED: Belinostat is a hydroxamate class HDAC inhibitor that has demonstrated activity in peripheral T-cell lymphoma and is undergoing clinical trials for non-hematologic malignancies. We studied the pharmacokinetics of belinostat in hepatocellular carcinoma patients to determine the main pathway of metabolism of belinostat. The pharmacokinetics of belinostat in liver cancer patients were characterized by rapid plasma clearance of belinostat with extensive metabolism with more than 4-fold greater relative systemic exposure of major metabolite, belinostat glucuronide than that of belinostat. There was significant interindividual variability of belinostat glucuronidation. The major pathway of metabolism involves UGT1A1-mediated glucuronidation and a good correlation has been identified between belinostat glucuronide formation and glucuronidation of known UGT1A1 substrates. In addition, liver microsomes harboring UGT1A1*28 alleles have lower glucuronidation activity for belinostat compared to those with wildtype UGT1A1. The main metabolic pathway of belinostat is through glucuronidation mediated primarily by UGT1A1, a highly polymorphic enzyme. The clinical significance of this finding remains to be determined. TRIAL REGISTRATION: ClinicalTrials.gov NCT00321594.


Assuntos
Antineoplásicos/farmacocinética , Carcinoma Hepatocelular/metabolismo , Glucuronosiltransferase/metabolismo , Inibidores de Histona Desacetilases/farmacocinética , Ácidos Hidroxâmicos/farmacocinética , Neoplasias Hepáticas/metabolismo , Redes e Vias Metabólicas , Sulfonamidas/farmacocinética , Antineoplásicos/metabolismo , Antineoplásicos/toxicidade , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/genética , Estabilidade de Medicamentos , Expressão Gênica , Genótipo , Glucuronosiltransferase/genética , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/toxicidade , Humanos , Concentração de Íons de Hidrogênio , Ácidos Hidroxâmicos/metabolismo , Ácidos Hidroxâmicos/toxicidade , Cinética , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/genética , Metaboloma , Microssomos Hepáticos/metabolismo , Especificidade por Substrato , Sulfonamidas/metabolismo , Sulfonamidas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...