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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 326(2): 657-71, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18451316

RESUMO

Methapyrilene (MP), a 2-thiophene H(1)-receptor antagonist, is a model toxicant in the genomic and proteomic analyses of hepatotoxicity. In rats, it causes an unusual periportal necrosis that is hypothetically attributed to chemically reactive and cytotoxic metabolites. We have characterized the bioactivation of MP by hepatic microsomes and primary rat hepatocytes, and we established a possible causal linkage with cytotoxicity. Methapyrilene tritiated at C-2 of the diaminoethane moiety ([3H]MP) was metabolized via an NADPH-dependent pathway to intermediates that combined irreversibly with microsomes (rat > mouse approximately human). This binding was attenuated by the cytochrome P450 (P450) inhibitor 1-aminobenzotriazole and thiols but not by trapping agents for iminium ions and aldehydes. Reactive intermediates were trapped as thioether adducts of monooxygenated MP. Mass spectrometric and hydrogen/deuterium exchange analysis of the glutathione adduct produced by rat liver microsomes indicated that the metabolite was most probably a thioether of MP S-oxide substituted in the thiophene ring. The glutathione adduct was formed by rat hepatocytes and eliminated in bile by rats administered [3H]MP intravenously. MP produced concentration- and time-dependent cytotoxicity, depleted glutathione, and underwent irreversible binding to the hepatocytes before a significant increase in cell damage was observed. P450 inhibitors reduced turnover of the drug, production of the glutathione adduct, irreversible binding, and cytotoxicity but inhibited glutathione depletion selectively. MP underwent lesser turnover and bioactivation in mouse hepatocytes and was not cytotoxic. Analogs with phenyl and p-methoxyphenyl rings were much less hepatocytotoxic than MP. Hepatotoxicity in rats was diminished by predosing with 1-aminobenzotriazole. For the first time, a thiophene ring substituent is identified as a bioactivation-dependent toxicophore in hepatocytes.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Hepatócitos/efeitos dos fármacos , Antagonistas dos Receptores Histamínicos H1 , Metapirileno , Microssomos Hepáticos/efeitos dos fármacos , Tiofenos/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Cromatografia Líquida de Alta Pressão , Glutationa/metabolismo , Hepatócitos/metabolismo , Antagonistas dos Receptores Histamínicos H1/química , Antagonistas dos Receptores Histamínicos H1/farmacocinética , Antagonistas dos Receptores Histamínicos H1/toxicidade , Humanos , Masculino , Metapirileno/química , Metapirileno/farmacocinética , Metapirileno/toxicidade , Camundongos , Camundongos Endogâmicos , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Ratos , Ratos Wistar , Tiofenos/química
2.
Clin Pharmacokinet ; 44(10): 1067-81, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16176119

RESUMO

BACKGROUND AND OBJECTIVES: Gefitinib (IRESSA, ZD1839), an epidermal growth factor receptor tyrosine kinase inhibitor, has been approved in several countries for the treatment of advanced non-small-cell lung cancer. Preclinical studies were conducted to determine the cytochrome P450 (CYP) isoenzymes involved in the metabolism of gefitinib and to evaluate the potential of gefitinib to cause drug interactions through inhibition of CYP isoenzymes. Based on these findings, three clinical studies were carried out to investigate pharmacokinetic drug interactions in vivo with gefitinib. METHODS: In preclinical studies radiolabelled gefitinib was incubated with: (i) hepatic microsomal protein in the presence of selective CYP inhibitors; and (ii) expressed CYP enzymes. Human hepatic microsomal protein was incubated with selective CYP substrates in the presence of gefitinib. Clinical studies were all phase I, open-label, single-centre studies; two had a randomised, two-way crossover design and the third was nonrandomised. The first and second studies investigated the pharmacokinetics of gefitinib in the presence of a potent CYP3A4 inducer (rifampicin [rifampin]) or inhibitor (itraconazole) in healthy male volunteers. The third study investigated the effects that gefitinib had on the pharmacokinetics of metoprolol, a CYP2D6 substrate, in patients with solid tumours. RESULTS: The results of preclinical studies demonstrated that CYP3A4 is involved in the metabolism of gefitinib and that gefitinib is a weak inhibitor of CYP2D6 activity. In clinical studies when gefitinib was administered in the presence of rifampicin, geometric mean (gmean) maximum concentration and area under the plasma concentration-time curve (AUC) were reduced by 65% and 83%, respectively; these changes were statistically significant. When gefitinib was administered in the presence of itraconazole, gmean AUC increased by 78% and 61% at gefitinib doses of 250 and 500 mg, respectively; these changes also being statistically significant. Coadministration of metoprolol with gefitinib resulted in a 35% increase in the metoprolol area under plasma concentration-time curve from time zero to the time of the last quantifiable concentration; this change was not statistically significant. There was no apparent change in the safety profile of gefitinib as a result of coadministration with other agents. CONCLUSIONS: Although CYP3A4 inducers may reduce exposure to gefitinib, further work is required to define any resultant effect on the efficacy of gefitinib. Exposure to gefitinib is increased by coadministration with CYP3A4 inhibitors, but since gefitinib is known to have a good tolerability profile, a dosage reduction is not recommended. Gefitinib is unlikely to exert a clinically relevant effect on the pharmacokinetics of drugs that are dependent on CYP2D6-mediated metabolism for their clearance, but the potential to increase plasma concentrations should be considered if gefitinib is coadministered with CYP2D6 substrates that have a narrow therapeutic index or are individually dose titrated.


Assuntos
Antineoplásicos/farmacocinética , Itraconazol/farmacologia , Metoprolol/farmacologia , Quinazolinas/farmacocinética , Rifampina/farmacologia , Adolescente , Adulto , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Estudos Cross-Over , Citocromo P-450 CYP2D6/metabolismo , Inibidores do Citocromo P-450 CYP2D6 , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/biossíntese , Combinação de Medicamentos , Interações Medicamentosas , Indução Enzimática , Inibidores Enzimáticos , Feminino , Gefitinibe , Humanos , Itraconazol/administração & dosagem , Itraconazol/farmacocinética , Masculino , Metoprolol/administração & dosagem , Metoprolol/farmacocinética , Microssomos Hepáticos/enzimologia , Pessoa de Meia-Idade , Neoplasias/tratamento farmacológico , Quinazolinas/administração & dosagem , Quinazolinas/uso terapêutico , Rifampina/administração & dosagem , Rifampina/farmacocinética
3.
J Med Chem ; 57(3): 970-86, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24422550

RESUMO

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) has been widely considered by the pharmaceutical industry as a target to treat metabolic syndrome in type II diabetics. We hypothesized that central nervous system (CNS) penetration might be required to see efficacy. Starting from a previously reported pyrimidine compound, we removed hydrogen-bond donors to yield 3, which had modest CNS penetration. More significant progress was achieved by changing the core to give 40, which combines good potency and CNS penetration. Compound 40 was dosed to diet-induced obese (DIO) mice and gave excellent target engagement in the liver and high free exposures of drug, both peripherally and in the CNS. However, no body weight reduction or effects on glucose or insulin were observed in this model. Similar data were obtained with a structurally diverse thiazole compound 51. This work casts doubt on the hypothesis that localized tissue modulation of 11ß-HSD1 activity alleviates metabolic syndrome.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , Adamantano/análogos & derivados , Adamantano/síntese química , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/síntese química , Síndrome Metabólica/tratamento farmacológico , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/química , Adamantano/farmacocinética , Adamantano/farmacologia , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Cristalografia por Raios X , Ciclopropanos/síntese química , Ciclopropanos/farmacocinética , Ciclopropanos/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Gorduras na Dieta/administração & dosagem , Humanos , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/farmacologia , Insulina/sangue , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Síndrome Metabólica/metabolismo , Síndrome Metabólica/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Modelos Moleculares , Pirazóis/síntese química , Pirazóis/farmacocinética , Pirazóis/farmacologia , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/farmacocinética , Tiazóis/farmacologia
4.
Endocrinology ; 154(12): 4580-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24169553

RESUMO

The enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) is a target for novel type 2 diabetes and obesity therapies based on the premise that lowering of tissue glucocorticoids will have positive effects on body weight, glycemic control, and insulin sensitivity. An 11ß-HSD1 inhibitor (compound C) inhibited liver 11ß-HSD1 by >90% but led to only small improvements in metabolic parameters in high-fat diet (HFD)-fed male C57BL/6J mice. A 4-fold higher concentration produced similar enzyme inhibition but, in addition, reduced body weight (17%), food intake (28%), and glucose (22%). We hypothesized that at the higher doses compound C might be accessing the brain. However, when we developed male brain-specific 11ß-HSD1 knockout mice and fed them the HFD, they had body weight and fat pad mass and glucose and insulin responses similar to those of HFD-fed Nestin-Cre controls. We then found that administration of compound C to male global 11ß-HSD1 knockout mice elicited improvements in metabolic parameters, suggesting "off-target" mechanisms. Based on the patent literature, we synthesized another 11ß-HSD1 inhibitor (MK-0916) from a different chemical series and showed that it too had similar off-target body weight and food intake effects at high doses. In summary, a significant component of the beneficial metabolic effects of these 11ß-HSD1 inhibitors occurs via 11ß-HSD1-independent pathways, and only limited efficacy is achievable from selective 11ß-HSD1 inhibition. These data challenge the concept that inhibition of 11ß-HSD1 is likely to produce a "step-change" treatment for diabetes and/or obesity.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Metabolismo Energético/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Triazóis/farmacologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Tecido Adiposo/metabolismo , Animais , Glicemia , Peso Corporal , Encéfalo/metabolismo , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/efeitos adversos , Relação Dose-Resposta a Droga , Feminino , Genótipo , Glucose/metabolismo , Hipoglicemiantes/química , Insulina/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Estrutura Molecular , Pirazóis/química , Pirimidinas/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Triazóis/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA