Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Xenobiotica ; 37(9): 972-85, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17896324

RESUMO

The in vitro metabolism of M445,526 (ZD6,126 phenol) was investigated by incubating [(14)C]-M445,526 at a concentration of 10 microg ml(-1) with human hepatic microsomes (4 mg ml(-1)) or human hepatocytes (2 x 10(6) cells ml(-1)) for up to 180 min. Following incubation with microsomes and hepatocytes, up to 78% and 40% of [(14)C]-M445,526 was metabolized after 180 and 120 min, respectively. High-performance liquid chromatography (HPLC) with radiochemical detection confirmed extensive metabolism of [(14)C]-M445,526 by microsomes and hepatocytes. Mass spectrometry and (1)H-NMR spectroscopy enabled structural identification of up to eight metabolites. Human liver microsomes formed one major (O-desmethyl) and three minor (a further O-desmethyl and two different hydroxylated) phase I metabolites. Human hepatocytes produced one major metabolite, a sulphate conjugate of the major O-desmethyl metabolite formed by microsomes. Four minor metabolites were also formed, primarily by O-demethylation with subsequent glucuronidation. Taken collectively, [(14)C]-M445,526 underwent extensive in vitro metabolism by human liver fractions. These data were confirmed by subsequent human in vivo studies.


Assuntos
Alcaloides/farmacocinética , Hepatócitos/metabolismo , Microssomos Hepáticos/metabolismo , Compostos Organofosforados/farmacocinética , Adulto , Idoso , Alcaloides/química , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacocinética , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacocinética , Cromatografia Líquida de Alta Pressão , Colchicina/análogos & derivados , Colchicina/química , Colchicina/farmacocinética , Feminino , Humanos , Técnicas In Vitro , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Compostos Organofosforados/química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacocinética
3.
Xenobiotica ; 36(1): 29-39, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16507511

RESUMO

Desmethyl-gefitinib is a major metabolite of gefitinib observed in human plasma at concentrations similar to those of gefitinib. The epidermal growth factor receptor (EGFR)-related inhibitory effects of gefitinib and desmethyl-gefitinib have been compared both in vitro, using enzyme kinase assays and tumour cell growth inhibition, and in vivo by assessment of tumour xenografts growth inhibition in the mouse. Both gefitinib (IC(50) = 0.022 microM) and its desmethyl metabolite (0.036 microM) inhibited subcellular EGFR tyrosine kinase activity with a similar potency and selectivity. However, desmethyl-gefitinib (IC(50) = 0.76 microM) was 15 times less active than gefitinib (0.049 microM) against EGF-stimulated KB cell growth in a whole cell assay. Following a preliminary pharmacokinetic study to compare apparent oral bioavailability, gefitinib (75 mg kg(-1)) and desmethyl-gefitinib (150 mg kg(-1)) were administered orally for 15 days to female nude mice bearing LoVo tumour xenografts. Tumour concentrations of gefitinib (AUC = 300 microg h g(-1)) were much higher than those of desmethyl-gefitinib (44.3 microg h g(-1)), although plasma concentrations of gefitinib (48.4 microg h ml(-1)) and desmethyl-gefitinib (39.0 microg h ml(-1)) were quite similar at these dose levels. Gefitinib produced significant tumour growth inhibition throughout the course of the study ultimately resulting in a 50% decrease (compared with controls) by day 15. In contrast, although present at comparable plasma levels, desmethyl-gefitinib had little effect on tumour growth and is, therefore, considered unlikely to contribute significantly to the therapeutic activity of gefitinib in the clinical situation.


Assuntos
Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Quinazolinas/administração & dosagem , Quinazolinas/sangue , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Gefitinibe , Humanos , Células KB , Dose Letal Mediana , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Distribuição Tecidual
4.
Xenobiotica ; 35(1): 39-50, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15788367

RESUMO

The in vitro metabolism of [(14)C]-gefitinib (1-3 microM) was investigated using human liver microsomes and a range of expressed human cytochrome P450 enzymes, with particular focus on the formation of O-desmethyl-gefitinib (M523595), the major metabolite observed in human plasma. High-performance liquid chromatography with ultraviolet light, radiochemical and mass spectral analysis, together with the availability of authentic standards, enabled quantification and structural identification of metabolites. On incubation with pooled human liver microsomes, [(14)C]-gefitinib underwent rapid and extensive metabolism to a number of metabolites, although M523595 was only a minor microsomal product. Formation of most metabolites was markedly decreased by ketoconazole, but M523595 production was inhibited only by quinidine. Gefitinib was metabolized extensively by expressed CYP3A4, producing a similar range of metabolites to liver microsomes, but M523595 was not formed. CYP1A2, 2C9 and 2C19 produced no measurable metabolism of gefitinib, while CYP3A5 produced a range of metabolites similar to CYP3A4, but to a much lower degree. In contrast, CYP2D6 catalysed rapid and extensive metabolism of gefitinib to M523595. While formation of M523595 was CYP2D6 mediated, the overall metabolism of gefitinib was dependent primarily on CYP3A4, and this was not obviously diminished in liver microsomes from CYP2D6 poor metabolizers.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Microssomos Hepáticos/metabolismo , Quinazolinas/farmacologia , Quinazolinas/farmacocinética , Células Cultivadas , Relação Dose-Resposta a Droga , Gefitinibe , Humanos , Microssomos Hepáticos/efeitos dos fármacos
5.
Xenobiotica ; 34(10): 901-15, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15764410

RESUMO

The pharmacokinetics of gefitinib and its metabolites in rat and dog were investigated in preclinical studies conducted to support the safety evaluation and clinical development of gefitinib, the first EGFR tyrosine kinase inhibitor approved for the treatment of non-small-cell lung cancer. Following intravenous dosing (5 mg kg(-1), gefitinib plasma half-life was 3-6h in rats and dogs, although studies using a more sensitive HPLC-MS assay produced longer estimates of half-life (7-14h). In these studies, plasma clearance was high (male rat: 25 ml min(-1) kg(-1); female rat: 16 ml min(-1) kg(-1); male dog: 16 ml min(-1) kg(-1)), as was the volume of distribution (8.0-10.41 kg(-1) in rat; 6.31 kg(-1) in dog), and exposure in female rats was double that in males. Following administration of [14C]-gefitinib, concentrations of radioactivity in plasma exceeded gefitinib throughout the profile, indicating the presence of circulating metabolites in both rat and dog. An HPLC-MS assay was developed to measure concentrations of gefitinib and five potential metabolites in plasma. All five metabolites were detected in the rat, but at levels much lower than gefitinib. In the dog, exposure to gefitinib and M523595 was similar, with much lower concentrations of M537194 and only trace levels of the other metabolites. This profile of metabolites is similar to that observed in man.


Assuntos
Receptores ErbB/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/sangue , Quinazolinas/farmacocinética , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Gefitinibe , Injeções Intravenosas , Masculino , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/sangue , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/administração & dosagem , Ratos , Ratos Wistar , Fatores Sexuais , Especificidade da Espécie
6.
Xenobiotica ; 34(10): 917-34, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15764411

RESUMO

Following oral administration of [14C]-gefitinib to albino and pigmented rats, radioactivity was widely and rapidly distributed, with the highest levels being found in liver, kidney, lung and gastrointestinal tract, but with only low levels penetrating the brain. Levels of radioactivity persisted in melanin-containing tissues (pigmented eye and skin). Binding to plasma proteins was high (86-94%) across the range of species examined and was 91% in human plasma. Substantial binding occurred to both human serum albumin and alpha-1 acid glycoprotein. Following oral and intravenous administration of [14C]-gefitinib, excretion of radioactivity by rat, dog and human occurred predominantly via the bile into faeces, with < 7% of the dose being eliminated in urine. In all three species, gefitinib was cleared primarily by metabolism. In rat, morpholine ring oxidation was the major route of metabolism, leading to the formation of M537194 and M608236 as the main biliary metabolites. Morpholine ring oxidation, together with production of M523595 by O-demethylation of the quinazoline moiety, were the predominant pathways in dog, with oxidative defluorination also occurring to a lesser degree. Pathways in healthy human volunteers were similar to dog, with O-demethylation and morpholine ring oxidation representing the major routes of metabolism.


Assuntos
Receptores ErbB/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacocinética , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Gefitinibe , Humanos , Masculino , Taxa de Depuração Metabólica , Especificidade de Órgãos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/sangue , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/administração & dosagem , Quinazolinas/sangue , Quinazolinas/urina , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Fatores Sexuais , Especificidade da Espécie , Distribuição Tecidual
7.
Xenobiotica ; 34(11-12): 983-1000, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15801543

RESUMO

The in vitro metabolism of gefitinib was investigated by incubating [14C]-gefitinib, as well as M537194, M387783 and M523595 (the main metabolites of gefitinib observed in man), at a concentration of 100 microM with human liver microsomes (4 mg ml(-1)) for 120 min. These relatively high substrate and microsomal protein concentrations were used in an effort to generate sufficient quantities of metabolites for identification. HPLC with ultraviolet light, radiochemical and mass spectral analysis, together with the availability of authentic standards, enabled quantification and structural identification of a large number of metabolites. Although 16 metabolites were identified, metabolism was restricted to three regions of the molecule. The major pathway involved morpholine ring-opening and step-wise removal of the morpholine ring and propoxy side chain. O-demethylation of the quinazoline methoxy group was a quantitatively less important pathway, in contrast to the clinical situation, where O-desmethyl gefitinib (M523595) is the predominant plasma metabolite. The third metabolic route, oxidative defluorination, was only a minor route of metabolism. Some metabolites were formed by a combination of these processes, but no metabolism was observed in other parts of the molecule. Incubation of gefitinib produced ten identified metabolites, but the use of the three main in vivo metabolites as additional substrates enabled a more comprehensive metabolic pathway to be constructed and this has been valuable in supporting the more limited data available from the human in vivo study.


Assuntos
Microssomos Hepáticos/metabolismo , Quinazolinas/farmacocinética , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Gefitinibe , Humanos , Taxa de Depuração Metabólica , Microssomos Hepáticos/efeitos dos fármacos , Quinazolinas/administração & dosagem
8.
Eur J Clin Nutr ; 57(10): 1235-41, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14506483

RESUMO

OBJECTIVE: To explore the feasibility of low-fat spreads as vehicles for folic acid (FA) fortification by determining the acute absorption of FA from a fortified spread. DESIGN: Double blind, crossover study to test each of the following treatments administered at 1-weekly intervals: (A) 20 g low-fat (40%) spread fortified with 200 microg FA and a placebo tablet; (B) 20 g low-fat placebo spread and a 200 microg FA tablet; (C) 20 g low-fat placebo spread and a placebo tablet. SUBJECTS: A total of 13 male volunteers, aged 31.8+/-13.2 y. MAIN OUTCOME MEASURES: Plasma total folate concentrations, measured before and up to 10 h after each treatment (n=10 samples per treatment). RESULTS: Plasma folate concentrations were significantly increased compared with baseline values 1 h after administration of the FA tablet, and 1.5 h after the FA spread, and remained significantly higher than the baseline values for up to 7 h after both treatments. The maximum plasma folate response (R(max)), corrected for baseline values and 'placebo response', was established between 1 and 3 h postprandially in response to both FA spread and FA tablet, and no significant difference in R(max) was found between the two treatments (13.4 vs 14.4 nmol/l, P=0.9). The acute absorption of FA from fortified spread relative to that from the tablet, calculated on the basis of area under the plasma folate response curve, was 67% (P=0.04). CONCLUSION: The absorption of FA from fortified low-fat spread, although lower than from a tablet, is effective. These results suggest that low-fat spreads, typically associated with fat-soluble vitamin fortification, may also be considered feasible as vehicles for FA fortification.


Assuntos
Suplementos Nutricionais , Ácido Fólico/sangue , Ácido Fólico/farmacocinética , Alimentos Fortificados , Adulto , Área Sob a Curva , Estudos Cross-Over , Dieta com Restrição de Gorduras , Método Duplo-Cego , Ácido Fólico/administração & dosagem , Humanos , Absorção Intestinal , Masculino , Comprimidos
9.
Xenobiotica ; 29(8): 847-57, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10553725

RESUMO

1. Zolmitriptan was extensively metabolized by freshly isolated human hepatocytes to a number of components including the three main metabolites observed in vivo (N-desmethyl-zolmitriptan, zolmitriptan N-oxide and the indole acetic acid derivative). In contrast, metabolism of zolmitriptan by human hepatic microsomes was extremely limited with only small amounts of the N-desmethyl and indole ethyl alcohol metabolites being produced. 2. Furafylline, a selective inhibitor of CYP1A2, almost completely abolished the hepatocellular metabolism of zolmitriptan and markedly inhibited formation of the N-desmethyl metabolite in microsomes. Chemical inhibitors, selective against other major human cytochrome P450 (CYP2C9, 2C19, 2D6 and 3A4), had no obvious effects. In addition, expressed human CYP1A2 was the only cytochrome P450 to form the N-desmethyl metabolite. 3. N-desmethyl-zolmitriptan was extensively metabolized by both human hepatocytes and microsomes. The indole acetic acid and ethyl alcohol derivatives were the major metabolites formed by hepatocytes, whereas only the indole ethyl alcohol derivative was produced by microsomes. Metabolism of N-desmethyl-zolmitriptan was not inhibited by cytochrome P450-selective chemical inhibitors nor was it observed following incubation with expressed human cytochrome P450. Clorgyline, a selective inhibitor of monoamine oxidase A (MAO-A), markedly inhibited the microsomal formation of the indole ethyl alcohol derivative. 4. Primary metabolism of zolmitriptan is dependent mainly on CYP1A2, whereas MAO-A is responsible for further metabolism of N-desmethyl-zolmitriptan, the active metabolite. Since the in vivo clearance of zolmitriptan is primarily dependent on metabolism, interactions with drugs that induce or inhibit CYP1A2 or MAO-A may be anticipated.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/metabolismo , Oxazóis/metabolismo , Oxazolidinonas , Agonistas do Receptor de Serotonina/metabolismo , Cimetidina/farmacologia , Clorgilina/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/genética , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Isoenzimas , Cetoconazol/farmacologia , Fígado/citologia , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Omeprazol/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Teofilina/análogos & derivados , Teofilina/farmacologia , Triptaminas
10.
J Oral Pathol Med ; 28(8): 360-3, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10478961

RESUMO

The antioxidant status of an individual is thought to be important in the development of potentially malignant oral lesions (PMOL) and oral squamous cell carcinoma (OSCC). To date, little detailed information on mucosal antioxidant status is available in a United Kingdom population and neither has the relationship between smoking and mucosal antioxidant status been established. Furthermore, it has been implied that serum levels of antioxidants and tissue levels in the oral mucosa should be equivalent, but that is unproven. To address these deficiencies in our knowledge we studied 60 individuals, all of whom had an oral mucosal biopsy and simultaneous venous blood sampling. Antioxidant levels were measured using high performance liquid chromatography. Smokers (n= 19) were found to have significantly lower levels of plasma beta-carotene (P<0.05) and significantly lower levels of tissue alpha-carotene (P<0.05) than non-smokers (n=41). Tissue alpha-carotene correlated with plasma levels, but this was not the case with alpha-tocopherol, retinol, lycopene or beta-carotene. This is the first data on oral mucosal antioxidant levels and provides baseline data from which to study patients with potentially malignant oral lesions and oral squamous cell carcinoma.


Assuntos
Antioxidantes/metabolismo , Mucosa Bucal/metabolismo , Fumar/metabolismo , Adolescente , Adulto , Carotenoides/sangue , Carotenoides/metabolismo , Dieta , Feminino , Frutas , Humanos , Masculino , Pessoa de Meia-Idade , Valores de Referência , Fumar/sangue , Verduras , beta Caroteno/sangue , beta Caroteno/metabolismo
11.
Xenobiotica ; 29(4): 395-408, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10375008

RESUMO

1. D0870, an azole antifungal agent, produced dose-related increases in total cytochrome P450 and aldrin epoxidase when administered as 14 daily oral doses (0, 0.5, 2.5 and 12.5 mg/kg/day) to the male rat. Administered as single doses, D0870 increased pentobarbitone-sleeping time in a dose-related manner. 2. In human hepatic microsomal incubations, D0870 produced pronounced inhibition of CYP2C9 (tolbutamide hydroxylase) and, to a lesser degree, CYP3A4 (testosterone 6beta-hydroxylase), but had more limited effects on CYP1A2, 2C19 and 2D6 activity. In comparison with ketoconazole, itraconazole and fluconazole, D0870 was the most potent inhibitor of CYP2C9 activity. It is predicted that D0870 may inhibit the in vivo clearance of CYP2C9 substrates by approximately 58%, thereby increasing their steady-state concentrations by 2.4 times, which would be of clinical significance for some compounds. 3. During incubation of [14C]-D0870 with cultured human hepatocytes for up to 72 h, two discrete metabolites (A and B) were formed. Formation of metabolite A was abolished by both quinidine and ketoconazole and is probably CYP3A4-mediated, whereas generation of metabolite B did not appear to be dependent on cytochrome P450. 4. D0870 has potential to produce both induction and inhibition of cytochrome P450 enzymes in man.


Assuntos
Antifúngicos/farmacologia , Interações Medicamentosas , Triazóis/farmacologia , Animais , Antifúngicos/administração & dosagem , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Feminino , Humanos , Hipnóticos e Sedativos/farmacologia , Concentração Inibidora 50 , Cinética , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Modelos Químicos , Pentobarbital/farmacologia , Ratos , Fatores de Tempo , Triazóis/administração & dosagem
12.
Xenobiotica ; 28(9): 845-53, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9764927

RESUMO

1. The potential of propofol to inhibit the activity of major human cytochrome P450 enzymes has been examined in vitro using human liver microsomes. Propofol produced inhibition of CYP1A2 (phenacetin O-deethylation), CYP2C9 (tolbutamide 4'-hydroxylation), CYP2D6 (dextromethorphan O-demethylation) and CYP3A4 (testosterone 6beta-hydroxylation) activities with IC50 = 40, 49, 213 and 32 microM respectively. Ki for propofol against all of these enzymes with the exception of CYP2D6, where propofol showed little inhibitory activity, was 30, 30 and 19 microM respectively for CYPs 1A2, 2C9 and 3A4. 2. Furafylline, sulphaphenazole, quinidine and ketoconazole, known selective inhibitors of CYPs 1A2, 2C9, 2D6 and 3A4 respectively, were much more potent than propofol having IC50 = 0.8, 0.5, 0.2 and 0.1 microM; furafylline and sulphaphenazole yielded Ki = 0.6 and 0.7 microM respectively. 3. The therapeutic blood concentration of propofol (20 microM; 3-4 microg/ml) together with the in vitro Ki estimates for each of the major human P450 enzymes have been used to estimate the extent of cytochrome P450 inhibition, which may be produced in vivo by propofol. This in vitro-in vivo extrapolation indicates that the degree of inhibition of CYP1A2, 2C9 and 3A4 activity which could theoretically be produced in vivo by propofol is relatively low (40-51%); this is considered unlikely to have any pronounced clinical significance. 4. Although propofol has now been used in > 190 million people since its launch in 1986, there are only single reports of possible drug interactions between propofol and either alfentanil or warfarin. Consequently, it is difficult to conclude from either the published literature or the ZENECA safety database whether there is any evidence to indicate that propofol produces clinically significant drug interactions through inhibition of cytochrome P450-related drug metabolism.


Assuntos
Anestésicos Intravenosos , Hidrocarboneto de Aril Hidroxilases , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Microssomos Hepáticos/enzimologia , Propofol/farmacologia , Esteroide 16-alfa-Hidroxilase , Citocromo P-450 CYP1A2 , Inibidores do Citocromo P-450 CYP1A2 , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Feminino , Humanos , Masculino , Oxigenases de Função Mista/antagonistas & inibidores , Oxirredutases O-Desmetilantes/antagonistas & inibidores , Propofol/sangue , Esteroide Hidroxilases/antagonistas & inibidores
13.
Xenobiotica ; 28(5): 465-78, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9622849

RESUMO

1. Bicalutamide, a non-steroidal antiandrogen, produced dose-related increases in total cytochrome P450 (P450) and aldrin epoxidase, but had no effect on ethoxyresorufin O-deethylase, when administered for 10 weeks at 0, 25, 75 and 150 mg/kg/day to the male dog. 2. In the male and female mouse, bicalutamide, administered orally at 75 mg/kg/day for 3 months, produced marked induction of total P450, ethoxycoumarin O-deethylase, pentoxyresorufin O-dealkylase and aldrin epoxidase. Immunoblotting showed that bicalutamide produced substantial induction of CYP2B isoforms, with lower increases in CYP3A. Immunohistochemistry of mouse liver sections also showed marked increases in the level of CYP2B isoforms, with an increase in the extent of distribution from centrilobular to panlobular; CYP3A isoforms were also increased, but to a lesser degree. 3. Bicalutamide, administered as 14 daily oral doses (250 mg/kg) to groups of male rats, produced increases primarily in ethoxycoumarin O-deethylase and erythromycin N-demethylase, together with smaller increases in ethoxyresorufin O-deethylase and pentoxyresorufin O-dealkylase; these changes were reversible within 7 days. Immunoblotting of microsomes and immunocytochemistry of liver sections showed that bicalutamide markedly induced CYP3A1, but had little effect on CYP2B1 in rat. Compared with dexamethasone, bicalutamide is a more selective inducer of CYP3A1 in rat. 4. Bicalutamide, administered to rats as 14 daily oral doses of 10 mg/kg, induced its own metabolism by stimulating both aromatic hydroxylation and direct glucuronidation. This effect was apparently offset by a concomitant decrease in hydrolysis of bicalutamide, resulting in no marked change in total amounts of dose eliminated over 2 days. 5. Although the secondary effects of enzyme induction result in thyroid hypertrophy and adenoma in rat and hepatocellular carcinoma in mouse following chronic administration of bicalutamide, these changes are considered to have little clinical relevance. In any case, bicalutamide does not produce enzyme induction in man at clinically relevant dose levels.


Assuntos
O-Dealquilase 7-Alcoxicumarina/biossíntese , Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Hidrocarboneto de Aril Hidroxilases , Citocromo P-450 CYP2B1/biossíntese , Sistema Enzimático do Citocromo P-450/biossíntese , Fígado/enzimologia , Microssomos Hepáticos/enzimologia , Oxigenases de Função Mista/biossíntese , Administração Oral , Antagonistas de Androgênios/administração & dosagem , Anilidas/administração & dosagem , Anilidas/farmacocinética , Animais , Bile/metabolismo , Células Cultivadas , Citocromo P-450 CYP3A , Cães , Indução Enzimática , Fezes/química , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/efeitos dos fármacos , NADPH-Ferri-Hemoproteína Redutase/biossíntese , Nitrilas , Ratos , Ratos Endogâmicos , Compostos de Tosil
14.
Xenobiotica ; 28(5): 493-506, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9622851

RESUMO

1. Although the importance of in vitro technology in supporting drug development is widely accepted, there is no real consensus about which approaches should be taken, which substrates should be used, or on the reliability and application of in vitro data. Consequently, as part of a collaborative project to characterize human liver with respect to the major forms of cytochrome P450, an interlaboratory comparison of the analysis of samples for form-specific activities was undertaken. 2. Microsomal fractions were isolated from five different human liver samples taken from the liver bank maintained at the Royal Postgraduate Medical School (RPMS). Aliquots from the five samples were sent to the 11 collaborating laboratories for characterization using their in-house, form-specific assays for cytochrome P450 activities. Although each laboratory assayed protein concentration, total cytochrome P450 content and enzyme activities were calculated using the protein estimation generated by RPMS to eliminate this possible source of variability. 3. With the exception of one laboratory, all estimates of protein concentration were similar (coefficient of variation, CoV, 9-13%) and the rank-order of the five samples was consistent across the laboratories. There was greater variability in the estimates of total cytochrome P450 content (CoV 28-43%), although again rank order of the samples across laboratories was fairly consistent. 4. The various laboratories used a number of different probe substrates, together with a range of conditions (substrate concentration, time of incubation, amount of protein), to assay for activity of CYP1A2, CYP2C9, CYP2D6, CYP2E1 and CYP3A4. However, apart from the occasional outlier, the five samples were ranked for activity of all these forms of cytochrome P450 with a high degree of consistency by the various laboratories and the choice of substrate had no appreciable effect on the ranking of the samples. 5. While this interlaboratory comparison has shown that greater consistency in the approach to in vivo determination of drug-metabolizing activity is desirable, there was little indication that any particular approach or substrate was superior to the others.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/metabolismo , Isoenzimas/metabolismo , Microssomos Hepáticos/enzimologia , Esteroide 16-alfa-Hidroxilase , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/análise , Humanos , Isoenzimas/análise , Laboratórios/normas , Oxigenases de Função Mista/metabolismo , Garantia da Qualidade dos Cuidados de Saúde , Reprodutibilidade dos Testes , Esteroide Hidroxilases/metabolismo , Especificidade por Substrato , Xenobióticos/farmacocinética
15.
J Pharm Pharmacol ; 48(6): 601-6, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8832494

RESUMO

4-(2-[7-Amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5] triazin-5-ylamino]ethyl)phenol (ZM 241385) is currently the most selective for the A2a adenosine receptor antagonist. This paper describes the in-vivo activity of ZM 241385 after administration by both oral and intraduodenal routes. In conscious spontaneously hypertensive rats, ZM 241385 (1-10 mg kg-1) selectively attenuated the mean arterial blood pressure response produced by exogenous adenosine (1 mg kg-1 min-1, i.v.) by up to 45% after oral administration. Activity of ZM 241385 was maintained for at least 6 h after 3 and 10 mg kg-1 (p.o.). In conscious normotensive cats, ZM 241385 attenuated the blood pressure responses to adenosine (0.6-1.0 mg kg-1, i.v.) by 94% after 10 mg kg-1 (p.o.) and by up to 74% after 0.3 mg kg-1 (i.v.). Duration of action of ZM 241385 up to 12 h (36% inhibition) was observed after 3 mg kg-1 (p.o.). In anaesthetized dogs and cats, ZM 241385, after intraduodenal administration (1-10 mg kg-1), produced a rapid (dose ratio 100-fold 15 min after administration of 10 mg kg-1 in the cat) and prolonged (dose ratio of 14 at 6 h after administration of 10 mg kg-1) attenuation of the vasodilatation responses to adenosine receptor stimulation. When administered by this route ZM 241385 was six times more potent than theophylline in the cat and at least twice as potent as theophylline in the dog. In conclusion, ZM 241385 is a potent, selective A2a adenosine receptor antagonist which is orally active, with a good duration of action by the enteric route in cat, rat and dog. It could therefore be used to evaluate the role of adenosine A2a receptors in the action of adenosine in-vivo.


Assuntos
Hemodinâmica/efeitos dos fármacos , Antagonistas de Receptores Purinérgicos P1 , Triazinas/farmacologia , Triazóis/farmacologia , Administração Oral , Animais , Gatos , Cães , Interações Medicamentosas , Feminino , Hipertensão/genética , Hipertensão/fisiopatologia , Injeções Intravenosas , Intubação Gastrointestinal , Masculino , Ratos , Especificidade da Espécie , Teofilina/antagonistas & inibidores , Teofilina/farmacologia , Triazinas/administração & dosagem , Triazinas/farmacocinética , Triazóis/administração & dosagem , Triazóis/farmacocinética , Vasodilatação/efeitos dos fármacos , Vasodilatadores/antagonistas & inibidores , Vasodilatadores/farmacologia
16.
Anticancer Drugs ; 7(1): 54-9, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8742099

RESUMO

The potential for hepatic enzyme induction by bicalutamide ('Casodex') was assessed in an open study in prostate cancer patients. A single, oral dose of antipyrine 1000 mg was given before and after 12 weeks' bicalutamide therapy [once daily 50 mg (n = 7) or 150 mg (n = 11)] and its pharmacokinetics and metabolism were determined. Plasma or saliva samples were taken for the measurement of antipyrine concentration. Urine samples were assayed for antipyrine and its three major metabolites. With bicalutamide 50 mg, plasma antipyrine concentrations were maximal between 2 and 4 h after administration, declined in a log-linear manner and were unaffected by bicalutamide therapy; with bicalutamide 150 mg, saliva antipyrine concentrations were maximal between 2 and 4 h, declined in a log-linear manner, and were also unaffected by bicalutamide therapy. Antipyrine half-life was 16.3% shorter after bicalutamide 50 mg (p < 0.05); a small decrease (13.5%) in half-life after bicalutamide 150 mg was not statistically significant. A small reduction (18.6%, p < 0.05) in the AUCinfinity for antipyrine was noted after bicalutamide 150 mg. A statistically significant reduction in antipyrine recovery was seen with the lower bicalutamide dose (23.7%, p < 0.05). The statistically significant changes were small in absolute terms and showed no dose-response relationship. Bicalutamide does not significantly induce the hepatic enzymes responsible for antipyrine metabolism and has no obvious potential for producing clinically significant drug interactions due to enzyme induction.


Assuntos
Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Fígado/efeitos dos fármacos , Neoplasias da Próstata/enzimologia , Idoso , Idoso de 80 Anos ou mais , Antagonistas de Androgênios/administração & dosagem , Anilidas/administração & dosagem , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/sangue , Anti-Inflamatórios não Esteroides/farmacocinética , Anti-Inflamatórios não Esteroides/urina , Antipirina/administração & dosagem , Antipirina/sangue , Antipirina/farmacocinética , Antipirina/urina , Indução Enzimática/efeitos dos fármacos , Humanos , Fígado/enzimologia , Masculino , Pessoa de Meia-Idade , Nitrilas , Neoplasias da Próstata/sangue , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/urina , Saliva/metabolismo , Compostos de Tosil
17.
FEBS Lett ; 368(3): 531-5, 1995 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-7635215

RESUMO

The motor protein non-claret disjunctional (ncd) moves towards the minus ends of microtubules (MTs), whereas its close relative kinesin moves in the opposite direction towards the plus ends of MTs. The mechanisms of movement and directional reversal for these motor proteins are unknown. Here we report the rate constants for MT activated ADP release from a recombinant double-headed ncd protein, GST-MC5, and a recombinant double-headed kinesin protein, K delta 401, measured using the fluorescent nucleotide analogues methylanthranilyol ATP (mantATP) and mantADP. Comparison of the maximal MT activated mantADP release rates for these proteins with their maximal MT activated mantATP turnover rates indicates that ADP release is the rate-limiting step for ATP turnover for both ncd and kinesin. This data supports the view that directional reversal may result from structural rather than chemical kinetic differences in the way the motors interact with MTs.


Assuntos
Difosfato de Adenosina/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Drosophila , Cinesinas/metabolismo , Proteínas dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/química , Animais , Ativação Enzimática , Corantes Fluorescentes/química , Glutationa Transferase/metabolismo , Cinética , Ligação Proteica , Suínos , ortoaminobenzoatos/química
18.
Xenobiotica ; 25(6): 623-33, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7483662

RESUMO

1. Casodex, a non-steroidal antiandrogen, is a racemic mixture of R-Casodex, the pharmacologically active (-)-enantiomer, and S-Casodex, the inactive (+)-enantiomer. Single oral doses of pseudo-racemic 14C-Casodex (10 mg/kg), prepared from mixtures of either 14C-labelled R-Casodex and unlabelled S-Casodex, or 14C-S-Casodex and unlabelled R-Casodex, were administered to the intact and bile duct-cannulated male rat. 2. Neither enantiomer underwent stereochemical inversion, but the pharmacokinetics of Casodex showed marked enantioselectivity. 3. After dosing R-labelled Casodex, plasma concentrations of R-Casodex increased slowly to reach a peak of 3.50 +/- 0.05 micrograms/ml (mean +/- SEM) at 12 h and, thereafter, declined monoexponentially with an elimination half-life of 24 h. Plasma concentrations of S-Casodex rose rapidly to reach a much lower peak of 0.97 +/- 0.06 microgram/ml at 3 h and, thereafter, declined rapidly, although there were insufficient data to determine the half-life. R-Casodex had a much higher AUC0-24 (66 micrograms.h/ml) than S-Casodex (12 micrograms.h/ml). Plasma drug concentrations measured using an achiral assay were in very good agreement with the sum of the enantiomer concentrations throughout the profile. R-Casodex comprised 94% of the total plasma radioactivity at 12 h, decreasing to 75% at 120 h. 4. Plasma concentration data generated after administration of S-Casodex were similar to those observed after dosing R-labelled Casodex. S-Casodex comprised about 74% of the total plasma radioactivity at 6 h and only 41% at 24 h. 5. The urine of intact animals contained 36 +/- 2 and 48 +/- 3% of the dose respectively up to 48 and 120 h after dosing with R-labelled Casodex, and 33 +/- 4 and 34 +/- 4% respectively after dosing with S-labelled Casodex. The urine and bile of the cannulated rat contained 43 +/- 2 and 21 +/- 2% of the dose respectively up to 48 h after dosing with R-labelled Casodex and 37 (n = 2) and 50% respectively after dosing with S-labelled Casodex. 6. After dosing with R- or S-labelled Casodex, the urinary radioactivity consisted of the carboxylic acid metabolite formed by hydrolytic cleavage at the amide, whereas biliary radioactivity consisted of hydroxy-Casodex and Casodex, mainly conjugated with glucuronic acid. The clearance of R-Casodex by each of these pathways of metabolism was less than that of S-Casodex, with direct glucuronidation and hydroxylation showing greater enantioselectivity than hydrolysis.


Assuntos
Antagonistas de Androgênios/metabolismo , Anilidas/metabolismo , Antagonistas de Androgênios/farmacocinética , Anilidas/sangue , Anilidas/farmacocinética , Animais , Fezes/química , Glucuronatos/metabolismo , Hidroxilação , Masculino , Nitrilas , Ratos , Estereoisomerismo , Compostos de Tosil
19.
J Muscle Res Cell Motil ; 15(3): 309-18, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7929796

RESUMO

2,3-butanedione 2-monoxime (BDM) inhibits muscle contraction and actomyosin ATPase both in fibres and in solution. It is potentially useful as a tool for exploring weak interactions between actin and myosin. We have examined the effect of BDM on several key steps of the myosin subfragment-1 and actomyosin subfragment-1 ATPase in solution. These studies show that BDM shifts the equilibrium between two actomyosin states towards a more weakly bound form when the acto.myosin complex has ADP alone or ADP and phosphate bound. We also confirm the findings of Herrmann and colleagues (1993, Biochemistry, 31, 12227-32) that the main effect of BDM on the myosin subfragment-1 ATPase is to slow the release of phosphate following ATP hydrolysis. Skinned fibre studies show that the effects of BDM and phosphate on the steady isometric tension of the fibres are additive. This is consistent with the interpretation that BDM is reducing fibre tension either by increasing phosphate binding or by a direct effect on the crossbridge. Tension transients induced by rapid pressure release were examined in single muscle fibres; they showed that BDM reduces the rate of tension generation following pressure release. This result suggest that BDM directly affects the force generating event in the crossbridge.


Assuntos
Actinas/análise , Actinas/metabolismo , Reativadores da Colinesterase/farmacologia , Diacetil/análogos & derivados , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Miosinas/análise , Miosinas/metabolismo , Difosfato de Adenosina/metabolismo , Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Diacetil/farmacologia , Músculo Esquelético/efeitos dos fármacos , Subfragmentos de Miosina/análise , Subfragmentos de Miosina/metabolismo , Fosfatos/farmacologia , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA