Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci Res ; 100(1): 329-338, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-32459013

RESUMO

Opioids are effective analgesics in the management of severe pain. However, tolerance, leading to dose escalation and adverse effects are significant limiting factors in their use. The role of peripheral opioid receptors in analgesia has been discussed especially under inflammatory conditions. The results from pharmacological and conditional knockout studies together do not provide a clear picture of the contribution of peripheral opioid receptors on antinociceptive tolerance and this needs to be evaluated. Therefore, we studied whether the peripherally restricted opioid receptor antagonist, methylnaltrexone (MNTX), could prevent morphine tolerance without attenuating the antinociceptive effect of morphine. Male Sprague-Dawley rats were treated for 7 days with increasing subcutaneous doses of morphine (5-30 mg/kg) and were coadministered saline, MNTX (0.5 or 2 mg/kg), or naltrexone (NTX; 2 mg/kg). Nociception was assessed with tail-flick, hotplate, and von Frey tests. Morphine, MNTX, and NTX concentrations in the plasma, brain, and spinal cord were measured by liquid chromatography-tandem mass spectrometry. In acute coadministration, NTX, but not MNTX, abolished the acute antinociceptive effects of morphine in all nociceptive tests. The antinociceptive tolerance after repeated morphine administration was also prevented by NTX but not by MNTX. MNTX penetrated to the spinal cord and the brain to some extent after repeated administration. The results do not support the use of MNTX for preventing opioid tolerance and also suggest that morphine tolerance is mediated by central rather than peripheral opioid receptors in the rat.


Assuntos
Morfina , Naltrexona , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Masculino , Morfina/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Compostos de Amônio Quaternário , Ratos , Ratos Sprague-Dawley , Receptores Opioides , Receptores Opioides mu
2.
Eur J Clin Pharmacol ; 74(12): 1615-1622, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30167757

RESUMO

PURPOSE: Buprenorphine has low oral bioavailability. Regardless of sublingual administration, a notable part of buprenorphine is exposed to extensive first-pass metabolism by the cytochrome P450 (CYP) 3A4. As drug interaction studies with buprenorphine are limited, we wanted to investigate the effect of voriconazole, a strong CYP3A4 inhibitor, on the pharmacokinetics and pharmacodynamics of oral buprenorphine. METHODS: Twelve healthy volunteers were given either placebo or voriconazole (orally, 400 mg twice on day 1 and 200 mg twice on days 2-5) for 5 days in a randomized, cross-over study. On day 5, they ingested 0.2 mg (3.6 mg during placebo phase) oral buprenorphine. We measured plasma and urine concentrations of buprenorphine and norbuprenorphine and monitored their pharmacological effects. Pharmacokinetic parameters were normalized for a buprenorphine dose of 1.0 mg. RESULTS: Voriconazole greatly increased the mean area under the plasma concentration-time curve (AUC0-18) of buprenorphine (4.3-fold, P < 0.001), its peak concentration (Cmax) (3.9-fold), half-life (P < 0.05), and excretion into urine (Ae; P < 0.001). Voriconazole also markedly enhanced the Cmax (P < 0.001), AUC0-18 (P < 0.001), and Ae (P < 0.05) of unconjugated norbuprenorphine but decreased its renal clearance (P < 0.001). Mild dizziness and nausea occurred during both study phases. CONCLUSIONS: Voriconazole greatly increases exposure to oral buprenorphine, mainly by inhibiting intestinal and liver CYP3A4. Effect on some transporters may explain elevated norbuprenorphine concentrations. Although oral buprenorphine is not commonly used, this interaction may become relevant in patients receiving sublingual buprenorphine together with voriconazole or other CYP3A4 or transporter inhibitors.


Assuntos
Analgésicos Opioides/farmacocinética , Antifúngicos/farmacologia , Buprenorfina/farmacocinética , Voriconazol/farmacologia , Adolescente , Adulto , Analgésicos Opioides/efeitos adversos , Antifúngicos/efeitos adversos , Área Sob a Curva , Biotransformação , Buprenorfina/efeitos adversos , Buprenorfina/análogos & derivados , Buprenorfina/metabolismo , Estudos Cross-Over , Citocromo P-450 CYP3A/metabolismo , Tontura/induzido quimicamente , Interações Medicamentosas , Feminino , Meia-Vida , Voluntários Saudáveis , Humanos , Masculino , Voriconazol/efeitos adversos , Adulto Jovem
3.
Basic Clin Pharmacol Toxicol ; 120(1): 38-45, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27312359

RESUMO

Spironolactone, eplerenone, chlorothiazide and furosemide are diuretics that have been suggested to have antinociceptive properties, for example via mineralocorticoid receptor antagonism. In co-administration, diuretics might enhance the antinociceptive effect of opioids via pharmacodynamic and pharmacokinetic mechanisms. Effects of spironolactone (100 mg/kg, i.p.), eplerenone (100 mg/kg, i.p.), chlorothiazide (50 mg/kg, i.p.) and furosemide (100 mg/kg, i.p.) were studied on acute oxycodone (0.75 mg/kg, s.c.)- and morphine (3 mg/kg, s.c.)-induced antinociception using tail-flick and hot plate tests in male Sprague Dawley rats. The diuretics were administered 30 min. before the opioids, and behavioural tests were performed 30 and 90 min. after the opioids. Concentrations of oxycodone, morphine and their major metabolites in plasma and brain were quantified by mass spectrometry. In the hot plate test at 30 and 90 min., spironolactone significantly enhanced the antinociceptive effect (% of maximum possible effect) of oxycodone from 10% to 78% and from 0% to 50%, respectively, and that of morphine from 12% to 73% and from 4% to 83%, respectively. The brain oxycodone and morphine concentrations were significantly increased at 30 min. (oxycodone, 46%) and at 90 min. (morphine, 190%). We did not detect any independent antinociceptive effects with the diuretics. Eplerenone and chlorothiazide did not enhance the antinociceptive effect of either opioid. The results suggest that spironolactone enhances the antinociceptive effect of both oxycodone and morphine by increasing their concentrations in the central nervous system.


Assuntos
Analgésicos/uso terapêutico , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Diuréticos/uso terapêutico , Neurônios/efeitos dos fármacos , Dor/prevenção & controle , Espironolactona/uso terapêutico , Analgésicos Opioides/sangue , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Clorotiazida/uso terapêutico , Interações Medicamentosas , Quimioterapia Combinada , Eplerenona , Furosemida/uso terapêutico , Masculino , Morfina/sangue , Morfina/metabolismo , Morfina/farmacocinética , Morfina/uso terapêutico , Neurônios/metabolismo , Oxicodona/sangue , Oxicodona/metabolismo , Oxicodona/farmacocinética , Oxicodona/uso terapêutico , Dor/sangue , Dor/metabolismo , Ratos Sprague-Dawley , Espironolactona/análogos & derivados , Distribuição Tecidual
4.
Eur J Clin Pharmacol ; 72(11): 1363-1371, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27510521

RESUMO

PURPOSE: This study aimed to determine possible effects of voriconazole and posaconazole on the pharmacokinetics and pharmacological effects of sublingual buprenorphine. METHODS: We used a randomized, placebo-controlled crossover study design with 12 healthy male volunteers. Subjects were given a dose of 0.4 mg (0.6 mg during placebo phase) sublingual buprenorphine after a 5-day oral pretreatment with either (i) placebo, (ii) voriconazole 400 mg twice daily on the first day and 200 mg twice daily thereafter or (iii) posaconazole 400 mg twice daily. Plasma and urine concentrations of buprenorphine and its primary active metabolite norbuprenorphine were monitored over 18 h and pharmacological effects were measured. RESULTS: Compared to placebo, voriconazole increased the mean area under the plasma concentration-time curve (AUC0-∞) of buprenorphine 1.80-fold (90 % confidence interval 1.45-2.24; P < 0.001), its peak concentration (Cmax) 1.37-fold (P < 0.013) and half-life (t ½ ) 1.37-fold (P < 0.001). Posaconazole increased the AUC00-∞ of buprenorphine 1.25-fold (P < 0.001). Most of the plasma norbuprenorphine concentrations were below the limit of quantification (0.05 ng/ml). Voriconazole, unlike posaconazole, increased the urinary excretion of norbuprenorphine 1.58-fold (90 % confidence interval 1.18-2.12; P < 0.001) but there was no quantifiable parent buprenorphine in urine. Plasma buprenorphine concentrations correlated with the pharmacological effects, but the effects did not differ significantly between the phases. CONCLUSIONS: Voriconazole, and to a minor extent posaconazole, increase plasma exposure to sublingual buprenorphine, probably via inhibition of cytochrome P450 3 A and/or P-glycoprotein. Care should be exercised in the combined use of buprenorphine with triazole antimycotics, particularly with voriconazole, because their interaction can be of clinical importance.


Assuntos
Analgésicos Opioides/farmacocinética , Antifúngicos/farmacologia , Buprenorfina/farmacocinética , Triazóis/farmacologia , Voriconazol/farmacologia , Administração Sublingual , Adulto , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Buprenorfina/efeitos adversos , Buprenorfina/farmacologia , Estudos Cross-Over , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Voluntários Saudáveis , Humanos , Masculino , Dor/tratamento farmacológico , Método Simples-Cego , Adulto Jovem
5.
Pharmacol Res Perspect ; 4(6): e00271, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-28097004

RESUMO

Buprenorphine is mainly metabolized by the cytochrome P450 (CYP) 3A4 enzyme. The aim of this study was to evaluate the role of first-pass metabolism in the interaction of rifampicin and analgesic doses of buprenorphine. A four-session paired cross-over study design was used. Twelve subjects ingested either 600 mg oral rifampicin or placebo once daily in a randomized order for 7 days. In the first part of the study, subjects were given 0.6-mg (placebo phase) or 0.8-mg (rifampicin phase) buprenorphine sublingually on day 7. In the second part of the study, subjects received 0.4-mg buprenorphine intravenously. Plasma concentrations of buprenorphine and urine concentrations of buprenorphine and its primary metabolite norbuprenorphine were measured over 18 h. Adverse effects were recorded. Rifampicin decreased the mean area under the dose-corrected plasma concentration-time curve (AUC 0-18) of sublingual buprenorphine by 25% (geometric mean ratio (GMR): 0.75; 90% confidence interval (CI) of GMR: 0.60, 0.93) and tended to decrease the bioavailability of sublingual buprenorphine, from 22% to 16% (P = 0.31). Plasma concentrations of intravenously administered buprenorphine were not influenced by rifampicin. The amount of norbuprenorphine excreted in the urine was decreased by 65% (P < 0.001) and 52% (P < 0.001) after sublingual and intravenous administration, respectively, by rifampicin. Adverse effects were frequent. Rifampicin decreases the exposure to sublingual but not intravenous buprenorphine. This can be mainly explained by an enhancement of CYP3A-mediated first-pass metabolism, which sublingual buprenorphine only partially bypasses. Concomitant use of rifampicin and low-dose sublingual buprenorphine may compromise the analgesic effect of buprenorphine.

6.
Drug Metab Dispos ; 41(1): 50-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23028140

RESUMO

Recent data suggest that the role of CYP3A4 in imatinib metabolism is smaller than presumed. This study aimed to evaluate the quantitative importance of different cytochrome P450 (P450) enzymes in imatinib pharmacokinetics. First, the metabolism of imatinib was investigated using recombinant P450 enzymes and human liver microsomes with P450 isoform-selective inhibitors. Thereafter, an in silico model for imatinib was constructed to perform pharmacokinetic simulations to assess the roles of P450 enzymes in imatinib elimination at clinically used imatinib doses. In vitro, CYP2C8 inhibitors and CYP3A4 inhibitors inhibited the depletion of 0.1 µM imatinib by 45 and 80%, respectively, and the formation of the main metabolite of imatinib, N-desmethylimatinib, by >50%. Likewise, recombinant CYP2C8 and CYP3A4 metabolized imatinib extensively, whereas other isoforms had minor effect on imatinib concentrations. In the beginning of imatinib treatment, the fractions of its hepatic clearance mediated by CYP2C8 and CYP3A4 were predicted to approximate 40 and 60%, respectively. During long-term treatment with imatinib 400 mg once or twice daily, up to 65 or 75% of its hepatic elimination was predicted to occur via CYP2C8, and only about 35 or 25% by CYP3A4, due to dose- and time-dependent autoinactivation of CYP3A4 by imatinib. Thus, although CYP2C8 and CYP3A4 are the main enzymes in imatinib metabolism in vitro, in silico predictions indicate that imatinib inhibits its own CYP3A4-mediated metabolism, assigning a key role for CYP2C8. During multiple dosing, pharmacogenetic polymorphisms and drug interactions affecting CYP2C8 activity may cause marked interindividual variation in the exposure and response to imatinib.


Assuntos
Antineoplásicos/farmacocinética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Benzamidas/farmacocinética , Inibidores do Citocromo P-450 CYP3A , Piperazinas/farmacocinética , Pirimidinas/farmacocinética , Antineoplásicos/sangue , Benzamidas/sangue , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP3A/metabolismo , Humanos , Mesilato de Imatinib , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Piperazinas/sangue , Pirimidinas/sangue , Proteínas Recombinantes/metabolismo
7.
Eur J Clin Pharmacol ; 68(5): 681-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22108774

RESUMO

PURPOSE: Zafirlukast is a substrate of cytochrome P450 2C9 (CYP2C9) and cytochrome P450 3A4 (CYP3A4) in vitro, but the role of these enzymes in its metabolism in vivo is unknown. To investigate the contribution of CYP2C9 and CYP3A4 to zafirlukast metabolism, we studied the effects of fluconazole and itraconazole on its pharmacokinetics (PK). METHODS: In a randomized crossover study, 12 healthy volunteers ingested fluconazole 200 mg (first dose 400 mg) once daily, itraconazole 100 mg (first dose 200 mg) twice daily, or placebo twice daily for 5 days, and on day 3, 20 mg zafirlukast. Plasma concentrations of zafirlukast and the antimycotics were measured up to 72 h. RESULTS: Fluconazole increased the total area under the plasma concentration-time curve (AUC) of zafirlukast 1.6-fold [95% confidence interval (CI) 1.3-2.0-fold, P < 0.001), and its peak plasma concentration 1.5-fold (95% CI 1.2-2.0-fold, P < 0.05). Fluconazole did not affect the time of peak plasma concentration or elimination half-life of zafirlukast. None of the zafirlukast PK variables differed significantly from the control in the itraconazole phase; e.g., the ratio to control of the total AUC of zafirlukast was 1.0 (95% CI 0.82-1.2) during the itraconazole phase. CONCLUSIONS: Fluconazole, but not itraconazole, increases zafirlukast plasma concentrations, strongly suggesting that CYP2C9 but not CYP3A4 participates in zafirlukast metabolism in humans.


Assuntos
Antifúngicos/farmacologia , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Inibidores do Citocromo P-450 CYP3A , Fluconazol/farmacologia , Itraconazol/farmacologia , Antagonistas de Leucotrienos/farmacocinética , Compostos de Tosil/farmacocinética , Adulto , Antiasmáticos/sangue , Antiasmáticos/farmacocinética , Antifúngicos/sangue , Antifúngicos/farmacocinética , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Biotransformação/efeitos dos fármacos , Estudos Cross-Over , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Feminino , Fluconazol/sangue , Fluconazol/farmacocinética , Estudos de Associação Genética , Meia-Vida , Humanos , Indóis , Itraconazol/análogos & derivados , Itraconazol/sangue , Itraconazol/farmacocinética , Antagonistas de Leucotrienos/sangue , Masculino , Fenilcarbamatos , Polimorfismo Genético , Sulfonamidas , Compostos de Tosil/sangue , Adulto Jovem
8.
Drug Metab Dispos ; 39(5): 904-11, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21289076

RESUMO

According to published in vitro studies, cytochrome P450 3A4 catalyzes montelukast 21-hydroxylation (M5 formation), whereas CYP2C9 catalyzes 36-hydroxylation (M6), the primary step in the main metabolic pathway of montelukast. However, montelukast is a selective competitive CYP2C8 inhibitor, and our recent in vivo studies suggest that CYP2C8 is involved in its metabolism. We therefore reevaluated the contributions of different cytochrome P450 (P450) enzymes, particularly that of CYP2C8, to the hepatic microsomal metabolism of montelukast using clinically relevant substrate concentrations in vitro. The effects of P450 isoform inhibitors on montelukast metabolism were examined using pooled human liver microsomes, and montelukast oxidations by human recombinant CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4, and CYP3A5 were investigated. The results verified the central role of CYP3A4 in M5 formation. The CYP2C8 inhibitors gemfibrozil 1-O-ß glucuronide and trimethoprim inhibited the depletion of 0.02 µM montelukast and formation of M6 from 0.05 µM montelukast more potently than did the CYP2C9 inhibitor sulfaphenazole. Likewise, recombinant CYP2C8 catalyzed montelukast depletion and M6 formation at a 6 times higher intrinsic clearance than did CYP2C9, whereas other P450 isoforms produced no M6. On the basis of depletion of 0.02 µM montelukast, CYP2C8 was estimated to account for 72% of the oxidative metabolism of montelukast in vivo, with a 16% contribution for CYP3A4 and 12% for CYP2C9. Moreover, CYP2C8 catalyzed the further metabolism of M6 more actively than did any other P450. In conclusion, CYP2C8 plays a major role in the main metabolic pathway of montelukast at clinically relevant montelukast concentrations in vitro.


Assuntos
Acetatos/metabolismo , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/metabolismo , Inibidores das Enzimas do Citocromo P-450 , Isoenzimas/antagonistas & inibidores , Antagonistas de Leucotrienos/metabolismo , Microssomos Hepáticos/enzimologia , Quinolinas/metabolismo , Acetatos/antagonistas & inibidores , Acetatos/farmacocinética , Ciclopropanos , Citocromo P-450 CYP2C8 , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Hidroxilação , Isoenzimas/metabolismo , Antagonistas de Leucotrienos/farmacocinética , Leucotrienos/agonistas , Microssomos Hepáticos/metabolismo , Modelos Teóricos , Oxirredução , Quinolinas/antagonistas & inibidores , Quinolinas/farmacocinética , Proteínas Recombinantes/metabolismo , Sulfetos , Fatores de Tempo
9.
Artigo em Inglês | MEDLINE | ID: mdl-19945922

RESUMO

We report a sensitive and robust method to determine cholic acid (CA), chenodeoxycholic acid (CDCA), deoxycholic acid (DCA), lithocholic acid (LCA), ursodeoxycholic acid (UDCA), and their taurine- and glycine-conjugate concentrations in human plasma using liquid chromatography-tandem mass spectrometry. Activated charcoal was utilized to prepare bile acid-free plasma, which served as the biological matrix for the preparation of standard and quality control samples. Plasma sample preparation involved solid-phase extraction. A total of 16 bile acids and 5 internal standards were separated on a reverse column by gradient elution and detected by tandem mass spectrometry in negative ion mode. The calibration curve was linear for all the bile acids over a range of 0.005-5micromol/L. The extraction recoveries for all the analytes fell in the range of 88-101%. Intra-day and inter-day coefficients of variation were all below 10%. A stability test showed that all the bile acids were stable in plasma for at least 6h at room temperature, at least three freeze-thaw cycles, in the -70 degrees C or -20 degrees C freezer for 2 months, and also in the reconstitution solution at 8 degrees C for 48h. Comparison of the matrix effect of bile acid-free plasma with that of real plasma indicated that the charcoal purification procedure did not affect the properties of charcoal-purified plasma as calibration matrix. This method has been used to determine the bile acid concentrations in more than 300 plasma samples from healthy individuals. In conclusion, this method is suitable for the simultaneous quantification of individual bile acids in human plasma.


Assuntos
Ácidos Cólicos/sangue , Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas em Tandem/métodos , Carvão Vegetal , Ácidos Cólicos/química , Estabilidade de Medicamentos , Glicina/química , Humanos , Modelos Lineares , Padrões de Referência , Sensibilidade e Especificidade , Extração em Fase Sólida , Taurina/química , Temperatura
10.
Pharmacogenet Genomics ; 19(6): 447-57, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19387419

RESUMO

BACKGROUND AND OBJECTIVE: Organic anion transporting polypeptide 1B1 (OATP1B1, encoded by SLCO1B1) is a sinusoidal influx transporter of human hepatocytes. Our aim was to characterize the role of OATP1B1 in the hepatic uptake of bile acids in vivo. METHODS: Fasting blood samples were drawn from 24 healthy volunteers with SLCO1B1 c.388AA-c.521TT (*1A/*1A) genotype, eight with c.388GG-c.521TT (*1B/*1B) genotype, 24 with c.521TC genotype, and nine with c.521CC genotype. Plasma concentrations of 15 endogenous bile acids, their synthesis marker, and cholesterol were determined by liquid chromatography-tandem mass spectrometry. RESULTS: The concentrations of ursodeoxycholic acid, glycoursodeoxycholic acid, chenodeoxycholic acid, and glycochenodeoxycholic acid were approximately 50-240% higher in individuals with the SLCO1B1 c.521CC, c.521TC, or c.388AA-c.521TT genotype than in those with the c.388GG-c.521TT genotype (P<0.05), with the largest differences seen between the c.521CC and c.388GG-c.521TT individuals. The concentration of tauroursodeoxycholic acid was approximately 120% higher in individuals with the c.521TC genotype and that of taurochenodeoxycholic acid 110% higher in individuals with the c.521CC or c.521TC genotype than in those with the c.388GG-c.521TT genotype (P<0.05). The cholic acid concentration was approximately 30% higher in individuals with the c.521CC or c.388AA-c.521TT genotype than in those with the c.388GG-c.521TT genotype (P<0.05), but its conjugates remained unaffected by the genotype. The bile acid synthesis marker 7alpha-hydroxy-4-cholesten-3-one/cholesterol concentration ratio was 62 or 45% higher in the c.388AA-c.521TT participants than in the c.388GG-c.521TT or c.521TC participants, respectively (P<0.05). CONCLUSION: SLCO1B1 polymorphism considerably affects the disposition of several endogenous bile acids and bile acid synthesis marker, indicating that OATP1B1 plays an important role in the hepatic uptake of bile acids in vivo in humans.


Assuntos
Ácidos e Sais Biliares/sangue , Colestenonas/sangue , Transportadores de Ânions Orgânicos/genética , Polimorfismo Genético/genética , Adulto , Biomarcadores/sangue , Colesterol/sangue , Feminino , Genótipo , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado , Masculino , Transportadores de Ânions Orgânicos/metabolismo
11.
Br J Clin Pharmacol ; 62(3): 345-57, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16934051

RESUMO

AIMS: Case reports suggest an interaction between rofecoxib and the CYP1A2 substrate tizanidine. Our objectives were to explore the extent and mechanism of this possible interaction and to determine the CYP1A2 inhibitory potency of rofecoxib. METHODS: In a randomized, double-blind, two-phase cross-over study, nine healthy subjects took 25 mg rofecoxib or placebo daily for 4 days and, on day 4, each ingested 4 mg tizanidine. Plasma concentrations and the urinary excretion of tizanidine, its metabolites (M) and rofecoxib, and pharmacodynamic variables were measured up to 24 h. On day 3, a caffeine test was performed to estimate CYP1A2 activity. RESULTS: Rofecoxib increased the area under the plasma concentration-time curve (AUC(0-infinity)) of tizanidine by 13.6-fold [95% confidence interval (CI) 8.0, 15.6; P < 0.001), peak plasma concentration (C(max)) by 6.1-fold (4.8, 7.3; P < 0.001) and elimination half-life (t(1/2)) from 1.6 to 3.0 h (P < 0.001). Consequently, rofecoxib markedly increased the blood pressure-lowering and sedative effects of tizanidine (P < 0.05). Rofecoxib increased several fold the tizanidine/M-3 and tizanidine/M-4 ratios in plasma and urine and the tizanidine/M-5, tizanidine/M-9 and tizanidine/M-10 ratios in urine (P < 0.05). In addition, it increased the plasma caffeine/paraxanthine ratio by 2.4-fold (95% CI 1.4, 3.4; P = 0.008) and this ratio correlated with the tizanidine/metabolite ratios. Finally, the AUC(0-25) of rofecoxib correlated with the placebo phase caffeine/paraxanthine ratio (r = 0.80, P = 0.01). CONCLUSIONS: Rofecoxib is a potent inhibitor of CYP1A2 and it greatly increases the plasma concentrations and adverse effects of tizanidine. The findings suggest that rofecoxib itself is also metabolized by CYP1A2, raising concerns about interactions between rofecoxib and other CYP1A2 substrate and inhibitor drugs.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Inibidores do Citocromo P-450 CYP1A2 , Lactonas/farmacologia , Sulfonas/farmacologia , Adulto , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Clonidina/análogos & derivados , Clonidina/farmacologia , Método Duplo-Cego , Interações Medicamentosas , Humanos , Masculino
12.
Basic Clin Pharmacol Toxicol ; 99(1): 44-51, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16867170

RESUMO

Our objective was to identify the cytochrome P450 (CYP) enzymes that metabolise pioglitazone and to examine the effects of the CYP2C8 inhibitors montelukast, zafirlukast, trimethoprim and gemfibrozil on pioglitazone metabolism in vitro. The effect of different CYP isoform inhibitors on the elimination of a clinically relevant concentration of pioglitazone (1 microM) and the formation of the main primary metabolite M-IV were studied using pooled human liver microsomes. The metabolism of pioglitazone by CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4 and CYP3A5 was investigated using human recombinant CYP isoforms. In particular, the inhibitors of CYP2C8, but also those of CYP3A4, markedly inhibited the elimination of pioglitazone and the formation of M-IV by HLM. Inhibitors selective to other CYP isoforms had a minor effect only. Of the recombinant isoforms, CYP2C8 (20 pmol/ml) metabolised pioglitazone markedly (56% in 60 min.), and also CYP3A4 had a significant effect (37% in 60 min.). Montelukast, zafirlukast, trimethoprim and gemfibrozil inhibited pioglitazone elimination in HLM with IC50 values of 0.51 microM, 1.0 microM, 99 microM and 98 microM, respectively, and the formation of the metabolite M-IV with IC50 values of 0.18 microM, 0.78 microM, 71 microM and 59 microM, respectively. In conclusion, pioglitazone is metabolised mainly by CYP2C8 and to a lesser extent by CYP3A4 in vitro. CYP2C9 is not significantly involved in the elimination of pioglitazone. The effect of different CYP2C8 inhibitors on pioglitazone pharmacokinetics needs to be evaluated also in vivo because, irrespective of their in vitro CYP2C8 inhibitory potency, their pharmacokinetic properties may affect the extent of interaction.


Assuntos
Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores Enzimáticos/farmacologia , Hipoglicemiantes/farmacocinética , Tiazolidinedionas/farmacocinética , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP3A , Interações Medicamentosas , Humanos , Hipoglicemiantes/farmacologia , Técnicas In Vitro , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , NADP/metabolismo , Pioglitazona , Tiazolidinedionas/farmacologia
13.
Basic Clin Pharmacol Toxicol ; 98(1): 79-85, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16433896

RESUMO

The effects of five HIV protease inhibitors (amprenavir, indinavir, nelfinavir, ritonavir and saquinavir) on cytochrome P450 (CYP) 3A4, 3A5 and 3A7 activities were studied in vitro using testosterone 6beta-hydroxylation in recombinant CYP3A4, CYP3A5 and CYP3A7 enzymes. The protease inhibitors showed differential inhibitory effects on the three CYP3A forms. Ritonavir and saquinavir were non-selective and preferential inhibitors of CYP3A4 and CYP3A5 (K(i) 0.03 microM and 0.6-0.8 microM for ritonavir and saquinavir, respectively), and weaker inhibitors of CYP3A7 (K(i) 0.6 microM and 1.8 microM, respectively). Nelfinavir was a potent and non-selective inhibitor of all three CYP3A forms (K(i) 0.3-0.4 microM). Amprenavir and indinavir preferentially inhibited CYP3A4 (K(i) 0.1 microM and 0.2 microM, respectively), with weaker inhibitory effects on CYP3A5 (K(i) 0.5 microM and 2.2 microM, respectively) and CYP3A7 (K(i) 2.1 microM and 10.6 microM, respectively). In conclusion, significant differences exist in the inhibitory potency of protease inhibitors for different CYP3A forms. Ritonavir, nelfinavir, saquinavir and amprenavir seem to be prone to drug-drug interactions by inhibiting both CYP3A4 and CYP3A5. Especially nelfinavir and ritonavir also have a potential to inhibit foetal CYP3A7-mediated drug metabolism and some endogenous pathways that may be crucial to normal foetal development, while indinavir has the lowest potential to inhibit CYP3A5 and CYP3A7.


Assuntos
Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Inibidores das Enzimas do Citocromo P-450 , Inibidores da Protease de HIV/farmacologia , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Carbamatos/farmacologia , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Furanos , Humanos , Hidroxilação , Técnicas In Vitro , Indinavir/farmacologia , Isoenzimas , Cinética , Nelfinavir/farmacologia , Proteínas Recombinantes , Ritonavir/farmacologia , Saquinavir/farmacologia , Sulfonamidas/farmacologia , Testosterona/metabolismo
14.
Br J Clin Pharmacol ; 61(1): 70-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16390353

RESUMO

AIMS: The effect of enzyme induction on the pharmacokinetics of pioglitazone, a thiazolidinedione antidiabetic drug that is metabolized primarily by CYP2C8, is not known. Rifampicin is a potent inducer of several CYP enzymes and our objective was to study its effects on the pharmacokinetics of pioglitazone in humans. METHODS: In a randomized, two-phase crossover study, ten healthy subjects ingested either 600 mg rifampicin or placebo once daily for 6 days. On the last day, they received a single oral dose of 30 mg pioglitazone. The plasma concentrations and cumulative excretion of pioglitazone and its active metabolites M-IV and M-III into urine were measured up to 48 h. RESULTS: Rifampicin decreased the mean total area under the plasma concentration-time curve (AUC(0-infinity)) of pioglitazone by 54% (range 20-66%; P = 0.0007; 95% confidence interval -78 to -30%) and shortened its dominant elimination half-life (t(1/2)) from 4.9 to 2.3 h (P = 0.0002). No significant effect on peak concentration (C(max)) or time to peak (t(max)) was observed. Rifampicin increased the apparent formation rate of M-IV and shortened its t(max) (P < 0.01). It also decreased the AUC(0-infinity) of M-IV (by 34%; P = 0.0055) and M-III (by 39%; P = 0.0026), shortened their t1/2 (M-IV by 50%; P = 0.0008, and M-III by 55%; P = 0.0016) and increased the AUC(0-infinity) ratios of M-IV and M-III to pioglitazone by 44% (P = 0.0011) and 32% (P = 0.0027), respectively. Rifampicin increased the M-IV/pioglitazone and M-III/pioglitazone ratios in urine by 98% (P = 0.0015) and 95% (P = 0.0024). A previously unrecognized metabolite M-XI, tentatively identified as a dihydroxy metabolite, was detected in urine during both phases, and rifampicin increased the ratio of M-XI to pioglitazone by 240% (P = 0.0020). CONCLUSIONS: Rifampicin caused a substantial decrease in the plasma concentration of pioglitazone, probably by induction of CYP2C8. Concomitant use of rifampicin with pioglitazone may decrease the efficacy of the latter drug.


Assuntos
Inibidores Enzimáticos/administração & dosagem , Hipoglicemiantes/farmacocinética , Rifampina/administração & dosagem , Tiazolidinedionas/farmacocinética , Administração Oral , Adulto , Área Sob a Curva , Estudos Cross-Over , Interações Medicamentosas , Indução Enzimática , Feminino , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/sangue , Masculino , Pioglitazona , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/sangue
15.
Clin Pharmacol Ther ; 78(4): 388-99, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16198658

RESUMO

BACKGROUND AND OBJECTIVE: Repaglinide is an antidiabetic drug metabolized by cytochrome P450 (CYP) 2 C 8 and 3A4, and it appears to be a substrate of the hepatic uptake transporter organic anion transporting polypeptide 1B1 (OATP1B1). We studied the effects of cyclosporine (INN, ciclosporin), an inhibitor of CYP3A4 and OATP1B1, on the pharmacokinetics and pharmacodynamics of repaglinide. METHODS: In a randomized crossover study, 12 healthy volunteers took 100 mg cyclosporine or placebo orally at 8 pm on day 1 and at 8 am on day 2. At 9 am on day 2, they ingested a single 0.25-mg dose of repaglinide. Concentrations of plasma and urine repaglinide and its metabolites (M), blood cyclosporine, and blood glucose were measured for 12 hours. The subjects were genotyped for single-nucleotide polymorphisms in CYP2C8, CYP3A5, SLCO1B1 (encoding OATP1B1), and ABCB1 (P-glycoprotein). The effect of cyclosporine on repaglinide metabolism was studied in human liver microsomes in vitro. RESULTS: During the cyclosporine phase, the mean peak repaglinide plasma concentration was 175% (range, 56%--365%; P=.013) and the total area under the plasma concentration-time curve [AUC0--infinity] was 244% (range, 119%--533%; P<.001) of that in the placebo phase. The amount of unchanged repaglinide and its metabolites M2 and M4 excreted in urine were raised 2.7--fold, 7.5--fold, and 5.0--fold, respectively, by cyclosporine (P<.001). The amount of M1 excreted in urine remained unchanged, but cyclosporine reduced the ratio of M1 to repaglinide by 62% (P<.001). Cyclosporine had no significant effect on the elimination half-life or renal clearance of repaglinide. Although the mean blood glucose-lowering effect of repaglinide was unaffected in this low-dose study with frequent carbohydrate intake, the subject with the greatest pharmacokinetic interaction had the greatest increase in blood glucose-lowering effect. The effect of cyclosporine on repaglinide AUC0-infinity was 42% lower in subjects with the SLCO1B1 521TC genotype than in subjects with the 521TT (reference) genotype (P=.047). In vitro, cyclosporine inhibited the formation of M1 (IC50 [concentration of inhibitor to cause 50% inhibition of original enzyme activity], 0.2 micromol/L) and M2 (IC50, 4.3 micromol/L) but had no effect on M4. CONCLUSIONS: Cyclosporine raised the plasma concentrations of repaglinide, probably by inhibiting its CYP3A4-catalyzed biotransformation and OATP1B1-mediated hepatic uptake. Coadministration of cyclosporine may enhance the blood glucose-lowering effect of repaglinide and increase the risk of hypoglycemia.


Assuntos
Carbamatos/sangue , Ciclosporina/farmacologia , Hipoglicemiantes/sangue , Imunossupressores/farmacologia , Piperidinas/sangue , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adulto , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Glicemia/efeitos dos fármacos , Carbamatos/farmacocinética , Carbamatos/farmacologia , Estudos Cross-Over , Ciclosporina/farmacocinética , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Sinergismo Farmacológico , Genótipo , Humanos , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/farmacologia , Imunossupressores/farmacocinética , Técnicas In Vitro , Transportador 1 de Ânion Orgânico Específico do Fígado , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Piperidinas/farmacocinética , Piperidinas/farmacologia , Polimorfismo de Nucleotídeo Único
16.
Clin Pharmacol Ther ; 78(4): 400-11, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16198659

RESUMO

BACKGROUND AND OBJECTIVE: Oral contraceptives (OCs) can inhibit drug metabolism, but their effect on various cytochrome P450 (CYP) enzymes and drugs can be different. Our objective was to study the effect of combined OCs, containing ethinyl estradiol (INN, ethinylestradiol) and gestodene, on CYP1A2 activity, as well as their interaction potential with tizanidine. METHODS: In a parallel-group study, 15 healthy women using OCs and 15 healthy women without OCs (control subjects) ingested a single dose of 4 mg tizanidine. Plasma and urine concentrations of tizanidine, as well as several of its metabolites (M-3, M-4, M-5, M-9, and M-10), and pharmacodynamic variables were measured until 24 hours after dosing. As a marker of CYP1A2 activity, an oral caffeine test was performed in both groups. RESULTS: The mean area under the plasma concentration-time curve from time 0 to infinity [AUC0-infinity] of tizanidine was 3.9 times greater (P<.001) and the mean peak plasma tizanidine concentration (Cmax) was 3.0 times higher (P<.001) in the OC users than in the control subjects. In 1 OC user the AUC0-infinity of tizanidine exceeded the mean AUC0-infinity of the control subjects by nearly 20 times. There were no significant differences in the elimination half-life or time to peak concentration in plasma of tizanidine between the groups. Tizanidine/metabolite ratios in plasma (M-3 and M-4) and urine (M-3, M-4, M-5, M-9, and M-10) were 2 to 10 times higher in the users of OCs than in the control subjects. In the OC group the excretion of unchanged tizanidine into urine was, on average, 3.8 times greater (P=.008) than in the control subjects. The plasma caffeine/paraxanthine ratio was 2.8 times higher (P<.001) in the OC users than in the control subjects. The caffeine/paraxanthine ratio correlated significantly with the AUC0-infinity and peak concentration of tizanidine in plasma, with its excretion into urine, and with, for example, the tizanidine/M-3 and tizanidine/M-4 area under the plasma concentration-time curve ratios. Both the systolic and diastolic blood pressures were lowered by tizanidine more in the OC users (-29+/- 10 mm Hg and -21+/- 8 mm Hg, respectively) than in the control subjects (-17+/- 9 mm Hg and -13+/- 5 mm Hg, respectively) (P < .01). CONCLUSIONS: OCs containing ethinyl estradiol and gestodene increase, to a clinically significant extent, the plasma concentrations and effects of tizanidine, probably mainly by inhibiting its CYP1A2-mediated presystemic metabolism. Care should be exercised when tizanidine is prescribed to OC users.


Assuntos
Agonistas alfa-Adrenérgicos/farmacocinética , Clonidina/análogos & derivados , Anticoncepcionais Orais Combinados/farmacologia , Inibidores do Citocromo P-450 CYP1A2 , Etinilestradiol/farmacologia , Norpregnenos/farmacologia , Adolescente , Agonistas alfa-Adrenérgicos/sangue , Agonistas alfa-Adrenérgicos/farmacologia , Adulto , Área Sob a Curva , Cafeína/sangue , Clonidina/sangue , Clonidina/farmacocinética , Clonidina/farmacologia , Sinergismo Farmacológico , Feminino , Meia-Vida , Humanos , Teofilina/sangue
17.
Basic Clin Pharmacol Toxicol ; 97(4): 249-56, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16176562

RESUMO

Repaglinide is an antidiabetic drug metabolised by cytochrome P450 (CYP) 2C8 and CYP3A4 enzymes. To clarify the mechanisms of observed repaglinide drug interactions, we determined the contribution of the two enzymes to repaglinide metabolism at different substrate concentrations, and examined the effect of fibrates and rifampicin on CYP2C8, CYP3A4 and repaglinide metabolism in vitro. We studied repaglinide metabolism using pooled human liver microsomes, recombinant CYP2C8 and recombinant CYP3A4 enzymes. The effect of quercetin and itraconazole on repaglinide metabolism, and of gemfibrozil, bezafibrate, fenofibrate and rifampicin on CYP2C8 (paclitaxel 6alpha-hydroxylation) and CYP3A4 (midazolam 1-hydroxylation) activities and repaglinide metabolism were studied using human liver microsomes. At therapeutic repaglinide concentrations (<0.4 microM), CYP2C8 and CYP3A4 metabolised repaglinide at similar rates. Quercetin (25 microM) and itraconazole (3 microM) inhibited the metabolism of 0.2 microM repaglinide by 58% and 71%, and that of 2 microM repaglinide by 56% and 59%, respectively. The three fibrates inhibited CYP2C8 (Ki: bezafibrate 9.7 microM, gemfibrozil 30.4 microM and fenofibrate 92.6 microM) and repaglinide metabolism (IC50: bezafibrate 37.7 microM, gemfibrozil 111 microM and fenofibrate 164 microM), but had no effect on CYP3A4. Rifampicin inhibited CYP2C8 (Ki 30.2 microM), CYP3A4 (Ki 18.5 microM) and repaglinide metabolism (IC50 13.7 microM). In conclusion, both CYP2C8 and CYP3A4 are important in the metabolism of therapeutic concentrations of repaglinide in vitro, but their predicted contributions in vivo are highly dependent on the scaling factor used. Gemfibrozil is only a moderate inhibitor of CYP2C8 and does not inhibit CYP3A4; inhibition of CYP-enzymes by parent gemfibrozil alone does not explain its interaction with repaglinide in vivo. Rifampicin competitively inhibits both CYP2C8 and CYP3A4, which can counteract its inducing effect in humans.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Carbamatos/farmacocinética , Sistema Enzimático do Citocromo P-450/metabolismo , Hipoglicemiantes/farmacocinética , Microssomos Hepáticos/efeitos dos fármacos , Piperidinas/farmacocinética , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Bezafibrato/farmacologia , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Interações Medicamentosas , Fenofibrato/farmacologia , Genfibrozila/farmacologia , Humanos , Técnicas In Vitro , Itraconazol/farmacologia , Microssomos Hepáticos/enzimologia , Midazolam/farmacologia , Paclitaxel/farmacologia , Quercetina/farmacologia , Rifampina/farmacologia
18.
Basic Clin Pharmacol Toxicol ; 97(2): 104-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15998357

RESUMO

Statins are involved in different types of drug interactions. Our objective was to study the effect of statins on cytochrome P450 (CYP) 2C8-mediated paclitaxel 6 alpha-hydroxylation by incubating paclitaxel and statins (0--100 microM) with pooled human liver microsomes. Simvastatin, lovastatin, atorvastatin and fluvastatin were the most potent inhibitors of CYP2C8 activity with K(i) (IC(50)) values of 7.1 (9.6) muM, 8.4 (15) microM, 16 (38) microM and 19 (37) microM, respectively. Cerivastatin, simvastatin acid and lovastatin acid were less potent inhibitors with K(i) (IC(50)) values ranging from 32 to 55 (30--67) microM. Rosuvastatin and pravastatin showed no appreciable effect on CYP2C8 activity even at 100 microM. In conclusion, all the statins tested, except rosuvastatin and pravastatin, had a significant inhibitory effect on the activity of CYP2C8 in vitro. Because many of the statins accumulate in the liver and because also their metabolites may inhibit CYP2C8 activity, in vivo studies are needed to investigate a possible interaction of simvastatin, lovastatin, atorvastatin and fluvastatin with CYP2C8 substrate drugs.


Assuntos
Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Algoritmos , Antineoplásicos Fitogênicos/metabolismo , Citocromo P-450 CYP2C8 , Humanos , Hidroxilação , Técnicas In Vitro , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Paclitaxel/metabolismo , Taxoides/farmacocinética
19.
Br J Clin Pharmacol ; 58(4): 390-6, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15373931

RESUMO

AIMS: Gemfibrozil markedly increases the plasma concentrations and blood glucose-lowering effects of repaglinide, but the effects of other fibrates on repaglinide pharmacokinetics are not known. Our aim was to investigate the effects of bezafibrate and fenofibrate on the pharmacokinetics and pharmacodynamics of repaglinide. METHODS: In a randomized, three-phase cross-over study, 12 healthy subjects received 400 mg bezafibrate, 200 mg fenofibrate or placebo once daily for 5 days. On day 5, a single 0.25 mg dose of repaglinide was ingested 1 h after the last pretreatment dose. The concentrations of plasma repaglinide, bezafibrate and fenofibrate and blood glucose were measured up to 7 h postdose. RESULTS: During the bezafibrate and fenofibrate phases, the total area under the concentration-time curve [AUC(0, infinity )] of repaglinide was 99% (95% confidence interval of the ratio to the control phase 73, 143%) and 99% (85, 127%) of the corresponding value during the placebo (control) phase, respectively. Bezafibrate and fenofibrate had no significant effect on the peak concentration (Cmax) of repaglinide. The mean half-life of repaglinide was 1.3 h in all phases. The blood glucose-lowering effect of repaglinide was not affected by bezafibrate or fenofibrate. The AUC(0,8 h) values for bezafibrate and fenofibrate varied 3.0-fold and 4.4-fold between individual subjects, respectively. Neither bezafibrate nor fenofibrate affected the pharmacokinetic variables of repaglinide. CONCLUSIONS: Bezafibrate and fenofibrate do not affect the pharmacokinetics or pharmacodynamics of repaglinide.


Assuntos
Bezafibrato/farmacologia , Carbamatos/farmacologia , Fenofibrato/farmacologia , Fenofibrato/farmacocinética , Hipolipemiantes/farmacologia , Hipolipemiantes/farmacocinética , Piperidinas/farmacologia , Adulto , Bezafibrato/administração & dosagem , Glicemia/metabolismo , Carbamatos/administração & dosagem , Estudos Cross-Over , Preparações de Ação Retardada , Interações Medicamentosas , Fenofibrato/administração & dosagem , Humanos , Hipolipemiantes/administração & dosagem , Masculino , Piperidinas/administração & dosagem , Comprimidos
20.
Br J Clin Pharmacol ; 57(3): 349-53, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14998432

RESUMO

AIMS: To identify the cytochrome p450 (CYP) enzyme(s) that catalyze the metabolism of tizanidine in vitro. METHODS: The effect of CYP isoform inhibitors on the elimination of tizanidine was studied using pooled human liver microsomes. The metabolism of the drug by a range of human recombinant CYP isoforms was then investigated. RESULTS: Incubation of tizanidine (80 nm) with human liver microsomes resulted in time- and NADPH-dependent substrate consumption with a half-life of 50 min, initial reaction velocity of 1.1 pmol x min-1 x mg-1 protein and intrinsic clearance of 17 ml x min-1 x kg-1. The predicted in vivo hepatic clearance (CLh) of tizanidine using the well-stirred and parallel-tube model was close (68% and 82%, respectively) to its estimated in vivo CLh. Fluvoxamine and furafylline strongly inhibited tizanidine metabolism. Inhibitors specific to isoforms other than CYP1A2 had no substantial effect. Recombinant CYP1A2 metabolized tizanidine to a substantial degree (35% in 45 min), but other recombinant CYPs had little metabolic capacity for the drug. CONCLUSIONS: CYP1A2 is primarily responsible for the metabolism of tizanidine. CYP1A2 inhibitors may inhibit its metabolism also in vivo.


Assuntos
Clonidina/análogos & derivados , Clonidina/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Inibidores do Citocromo P-450 CYP1A2 , Meia-Vida , Humanos , Microssomos Hepáticos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...