Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Hum Reprod ; 35(2): 257-264, 2020 02 29.
Artigo em Inglês | MEDLINE | ID: mdl-31958337

RESUMO

STUDY QUESTION: Does treatment of constitutional delay of growth and puberty (CDGP) in boys with aromatase inhibitor letrozole (Lz) or conventional low-dose testosterone (T) have differing effects on developing seminiferous epithelium? SUMMARY ANSWER: Anti-Müllerian hormone (AMH) declined similarly in both treatment groups, and the two Sertoli cell-derived markers (AMH and inhibin B (iB)) exhibited differing responses to changes in gonadotrophin milieu. WHAT IS KNOWN ALREADY: Boys with CDGP may benefit from puberty-inducing medication. Peroral Lz activates gonadotrophin secretion, whereas intramuscular low-dose T may transiently suppress gonadotrophins and iB. STUDY DESIGN, SIZE, DURATION: Sera of 28 boys with CDGP who participated in a randomised, controlled, open-label trial at four paediatric centres in Finland between August 2013 and January 2017 were analysed. The patients were randomly assigned to receive either Lz (2.5 mg/day) (n = 15) or T (1 mg/kg/month) (n = 13) for 6 months. PARTICIPANTS/MATERIALS, SETTING, METHODS: The 28 patients were at least 14 years of age, showed first signs of puberty, wanted medical attention for CDGP and were evaluated at 0, 3, 6 and 12 months of visits. AMH levels were measured with an electrochemiluminescence immunoassay and Lz levels with liquid chromatography coupled with tandem mass spectrometry. MAIN RESULTS AND THE ROLE OF CHANCE: AMH levels decreased in both treatment groups during the 12-month follow-up (P < 0.0001). Between 0 and 3 months, the changes in gonadotrophin levels (increase in the Lz group, decrease in the T group) correlated strongly with the changes in levels of iB (FSH vs iB, r = 0.55, P = 0.002; LH vs iB, r = 0.72, P < 0.0001), but not with the changes in AMH (P = NS). At 12 months, AMH levels did not differ between the groups (P = NS). Serum Lz levels (range, 124-1262 nmol/L) were largely explained by the Lz dose per weight (at 3 months r = 0.62, P = 0.01; at 6 months r = 0.52, P = 0.05). Lz levels did not associate with changes in indices of hypothalamic-pituitary-gonadal axis activity or Sertoli cell markers (in all, P = NS). LIMITATIONS, REASONS FOR CAUTION: The original trial was not blinded for practical reasons and included a limited number of participants. WIDER IMPLICATIONS OF THE FINDINGS: In early puberty, treatment-induced gonadotrophin stimulus was unable to counteract the androgen-mediated decrease in AMH, while changes in iB levels were associated with changes in gonadotrophin levels. AMH decreased similarly in both groups during the treatment, reassuring safety of developing seminiferous epithelium in both treatment approaches. Since a fixed dose of Lz induced variable serum Lz levels with a desired puberty-promoting effect in all boys, more research is needed to aim at a minimal efficient dose per weight. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the Academy of Finland, the Foundation for Pediatric Research, the Emil Aaltonen Foundation, Sigrid Juselius Foundation and Helsinki University Hospital Research Funds. The authors have nothing to disclose. TRIAL REGISTRATION NUMBER: NCT01797718.


Assuntos
Hormônio Antimülleriano/sangue , Transtornos do Crescimento/sangue , Inibinas/sangue , Letrozol/uso terapêutico , Puberdade Tardia/tratamento farmacológico , Testosterona/uso terapêutico , Adolescente , Biomarcadores/sangue , Criança , Feminino , Finlândia , Transtornos do Crescimento/tratamento farmacológico , Humanos , Hipogonadismo/sangue , Letrozol/administração & dosagem , Letrozol/sangue , Masculino , Puberdade Tardia/sangue , Testosterona/administração & dosagem
2.
Hum Exp Toxicol ; 36(4): 365-375, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27222493

RESUMO

Local anesthetic toxicity is thought to be mediated partly by inhibition of cardiac mitochondrial function. Intravenous (i.v.) lipid emulsion may overcome this energy depletion, but doses larger than currently recommended may be needed for rescue effect. In this randomized study with anesthetized pigs, we compared the effect of a large dose, 4 mL/kg, of i.v. 20% Intralipid® ( n = 7) with Ringer's acetate ( n = 6) on cardiovascular recovery after a cardiotoxic dose of bupivacaine. We also examined mitochondrial respiratory function in myocardial cell homogenates analyzed promptly after needle biopsies from the animals. Bupivacaine plasma concentrations were quantified from plasma samples. Arterial blood pressure recovered faster and systemic vascular resistance rose more rapidly after Intralipid than Ringer's acetate administration ( p < 0.0001), but Intralipid did not increase cardiac index or left ventricular ejection fraction. The lipid-based mitochondrial respiration was stimulated by approximately 30% after Intralipid ( p < 0.05) but unaffected by Ringer's acetate. The mean (standard deviation) area under the concentration-time curve (AUC) of total bupivacaine was greater after Intralipid (105.2 (13.6) mg·min/L) than after Ringer's acetate (88.1 (7.1) mg·min/L) ( p = 0.019). After Intralipid, the AUC of the lipid-un-entrapped bupivacaine portion (97.0 (14.5) mg·min/L) was 8% lower than that of total bupivacaine ( p < 0.0001). To conclude, 4 mL/kg of Intralipid expedited cardiovascular recovery from bupivacaine cardiotoxicity mainly by increasing systemic vascular resistance. The increased myocardial mitochondrial respiration and bupivacaine entrapment after Intralipid did not improve cardiac function.


Assuntos
Anestésicos Locais/toxicidade , Bupivacaína/toxicidade , Emulsões Gordurosas Intravenosas/farmacologia , Fosfolipídeos/farmacologia , Óleo de Soja/farmacologia , Anestésicos Locais/sangue , Animais , Bupivacaína/sangue , Respiração Celular/efeitos dos fármacos , Emulsões/farmacologia , Coração/efeitos dos fármacos , Coração/fisiologia , Hemodinâmica/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Suínos
3.
Clin Ther ; 38(10S): e1-e2, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27673600
4.
Anaesth Intensive Care ; 44(2): 270-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27029660

RESUMO

Intravenous lipid emulsion is, in some countries, the recommended treatment for local anaesthetic toxicity. Systemic local anaesthetic toxicity results in hypoxaemia and acidosis, and whether this influences the effects of lipid therapy on drug concentrations and cardiovascular recovery is currently unknown. Twenty anaesthetised pigs were given a 3-mg/kg bolus of levobupivacaine followed by a five minute phase of hypoventilation and 1 mmol/kg of lactic acid in one minute. After lactic acid infusion, pigs were treated, in randomised order, with either 20% lipid emulsion or Ringer's acetate for 30 min: a 1.5-ml/kg bolus followed by a 0.25-ml/kg/minute infusion. Haemodynamic parameters were recorded and blood samples were collected for pharmacokinetic analysis. There was no difference between the groups in the area under the plasma levobupivacaine concentration-time curve (AUC) or between that and AUC of unentrapped levobupivacaine in the Lipid group, or in the plasma half-lives. The cardiovascular outcome and normalisation of the electrocardiogram were similar in both groups. Five pigs developed marked hypotension: one in both groups died, while two in the Lipid group and one in the Ringer group needed adrenaline. Administration of lipid emulsion did not improve cardiovascular recovery from levobupivacaine toxicity exacerbated by acidosis and hypoxaemia. Lipid emulsion did not entrap levobupivacaine or affect levobupivacaine pharmacokinetics.


Assuntos
Acidose/tratamento farmacológico , Anestésicos Locais/intoxicação , Bupivacaína/análogos & derivados , Emulsões Gordurosas Intravenosas/uso terapêutico , Hipóxia/tratamento farmacológico , Animais , Bupivacaína/sangue , Bupivacaína/intoxicação , Dióxido de Carbono/sangue , Eletrocardiografia , Hemodinâmica , Levobupivacaína , Suínos
5.
Clin Pharmacol Ther ; 97(6): 650-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25704243

RESUMO

Carboxylesterase 1 (CES1) hydrolyzes the prodrug clopidogrel to an inactive carboxylic acid metabolite. We studied the pharmacokinetics and pharmacodynamics of 600 mg oral clopidogrel in healthy white volunteers, including 10 carriers and 12 noncarriers of CES1 c.428G>A (p.Gly143Glu, rs71647871) single nucleotide variation (SNV). Clopidogrel carboxylic acid to clopidogrel area under the plasma concentration-time curve from 0 hours to infinity (AUC0-∞ ) ratio was 53% less in CES1 c.428G>A carriers than in noncarriers (P = 0.009), indicating impaired hydrolysis of clopidogrel. Consequently, the AUC0-∞ of clopidogrel and its active metabolite were 123% (P = 0.004) and 67% (P = 0.009) larger in the c.428G>A carriers than in noncarriers. Consistent with these findings, the average inhibition of P2Y12 -mediated platelet aggregation 0-12 hours after clopidogrel intake was 19 percentage points higher in the c.428G>A carriers than in noncarriers (P = 0.036). In conclusion, the CES1 c.428G>A SNV increases clopidogrel active metabolite concentrations and antiplatelet effects by reducing clopidogrel hydrolysis to inactive metabolites.


Assuntos
Hidrolases de Éster Carboxílico/genética , Inibidores da Agregação Plaquetária/farmacologia , Polimorfismo de Nucleotídeo Único , Ticlopidina/análogos & derivados , Adulto , Clopidogrel , Feminino , Genótipo , Humanos , Hidrólise , Masculino , Ticlopidina/farmacocinética , Ticlopidina/farmacologia
6.
Clin Pharmacol Ther ; 96(4): 498-507, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24971633

RESUMO

Cerivastatin and repaglinide are substrates of cytochrome P450 (CYP)2C8, CYP3A4, and organic anion-transporting polypeptide (OATP)1B1. A recent study revealed an increased risk of rhabdomyolysis in patients using cerivastatin with clopidogrel, warranting further studies on clopidogrel interactions. In healthy volunteers, repaglinide area under the concentration-time curve (AUC(0-∞)) was increased 5.1-fold by a 300-mg loading dose of clopidogrel and 3.9-fold by continued administration of 75 mg clopidogrel daily. In vitro, we identified clopidogrel acyl-ß-D-glucuronide as a potent time-dependent inhibitor of CYP2C8. A physiologically based pharmacokinetic model indicated that inactivation of CYP2C8 by clopidogrel acyl-ß-D-glucuronide leads to uninterrupted 60-85% inhibition of CYP2C8 during daily clopidogrel treatment. Computational modeling resulted in docking of clopidogrel acyl-ß-D-glucuronide at the CYP2C8 active site with its thiophene moiety close to heme. The results indicate that clopidogrel is a strong CYP2C8 inhibitor via its acyl-ß-D-glucuronide and imply that glucuronide metabolites should be considered potential inhibitors of CYP enzymes.


Assuntos
Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Glucuronídeos/metabolismo , Inibidores da Agregação Plaquetária/metabolismo , Ticlopidina/análogos & derivados , Hidrocarboneto de Aril Hidroxilases/química , Carbamatos/farmacocinética , Domínio Catalítico , Clopidogrel , Simulação por Computador , Citocromo P-450 CYP2C8/química , Citocromo P-450 CYP3A/química , Interações Medicamentosas , Feminino , Humanos , Hipoglicemiantes/farmacocinética , Masculino , Desintoxicação Metabólica Fase II , Simulação de Acoplamento Molecular , Piperidinas/farmacocinética , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/farmacologia , Ticlopidina/química , Ticlopidina/metabolismo , Ticlopidina/farmacologia , Fatores de Tempo
7.
Clin Pharmacol Ther ; 95(3): 307-13, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24067745

RESUMO

Cytochrome P450 (CYP) enzymes, including CYP2C19 and CYP3A4, participate in the bioactivation of clopidogrel. Grapefruit juice constituents potently inactivate intestinal CYP3A4 and have been shown to inhibit CYP2C19 as well. In a randomized crossover study, 14 healthy volunteers ingested 200 ml of grapefruit juice or water three times daily for 3 days. On day 3, they ingested a single 600-mg dose of clopidogrel. Grapefruit juice reduced the peak plasma concentration (Cmax) of the active metabolite of clopidogrel to 13% of the control (range 11-17%, P < 0.001) and the area under the plasma concentration-time curve from 0 to 3 h to 14% (range 12-17%, P < 0.001) of the control, but it had no significant effect on the parent clopidogrel. Moreover, grapefruit juice markedly decreased the platelet-inhibitory effect of clopidogrel, as assessed with the VerifyNow P2Y12 test in two of the participants. In conclusion, concomitant use of grapefruit juice may impair the efficacy of clopidogrel. Therefore, the use of grapefruit juice is best avoided during clopidogrel therapy.


Assuntos
Bebidas/efeitos adversos , Biotransformação/efeitos dos fármacos , Citrus paradisi/efeitos adversos , Interações Alimento-Droga , Inibidores da Agregação Plaquetária/metabolismo , Ticlopidina/análogos & derivados , Adulto , Área Sob a Curva , Clopidogrel , Intervalos de Confiança , Estudos Cross-Over , Feminino , Genótipo , Meia-Vida , Humanos , Masculino , Testes de Função Plaquetária , Receptores Purinérgicos P2Y12/genética , Ticlopidina/metabolismo , Adulto Jovem
8.
Clin Pharmacol Ther ; 94(3): 383-93, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23657159

RESUMO

Cytochrome P450 (CYP) 3A4 is considered the most important enzyme in imatinib biotransformation. In a randomized, crossover study, 10 healthy subjects were administered gemfibrozil 600 mg or placebo twice daily for 6 days, and imatinib 200 mg on day 3, to study the significance of CYP2C8 in imatinib pharmacokinetics. Unexpectedly, gemfibrozil reduced the peak plasma concentration (Cmax) of imatinib by 35% (P < 0.001). Gemfibrozil also reduced the Cmax and area under the plasma concentration-time curve (AUC0-∞) of N-desmethylimatinib by 56 and 48% (P < 0.001), respectively, whereas the AUC0-∞ of imatinib was unaffected. Furthermore, gemfibrozil reduced the Cmax/plasma concentration at 24 h (C24 h) ratios of imatinib and N-desmethylimatinib by 44 and 17% (P < 0.05), suggesting diminished daily fluctuation of imatinib plasma concentrations during concomitant use with gemfibrozil. Our findings indicate significant participation of CYP2C8 in the metabolism of imatinib in humans, and support involvement of an intestinal influx transporter in imatinib absorption.


Assuntos
Antineoplásicos/farmacocinética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Benzamidas/farmacocinética , Genfibrozila/farmacologia , Hipolipemiantes/farmacologia , Piperazinas/farmacocinética , Pirimidinas/farmacocinética , Absorção , Adulto , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Hidrocarboneto de Aril Hidroxilases/genética , Benzamidas/administração & dosagem , Benzamidas/sangue , Estudos Cross-Over , Citocromo P-450 CYP2C8 , Antagonismo de Drogas , Feminino , Genótipo , Humanos , Mesilato de Imatinib , Masculino , Piperazinas/administração & dosagem , Piperazinas/sangue , Pirimidinas/administração & dosagem , Pirimidinas/sangue , Adulto Jovem
9.
Clin Pharmacol Ther ; 92(1): 68-71, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22588607

RESUMO

Bioactivation of the antiviral agent oseltamivir to active oseltamivir carboxylate is catalyzed by carboxylesterase 1 (CES1). After the screening of 860 healthy Finnish volunteers for the CES1 c.428G>A (p.Gly143Glu, rs121912777) polymorphism, a pharmacokinetic study with 75 mg oseltamivir was carried out in c.428G>A carriers and noncarriers. Heterozygous c.428GA carriers (n = 9) had 18% larger values of oseltamivir area under the plasma concentration-time curve from 0 h to infinity (AUC(0-∞)) (P = 0.025) and 23% smaller carboxylate-to-oseltamivir AUC(0-∞) ratio (P = 0.006) than noncarriers (n = 12). This shows that the CES1 c.428G>A polymorphism impairs oseltamivir bioactivation in humans.


Assuntos
Hidrolases de Éster Carboxílico , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Oseltamivir , Adulto , Antivirais/administração & dosagem , Antivirais/farmacocinética , Área Sob a Curva , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Relação Dose-Resposta a Droga , Monitoramento de Medicamentos , Feminino , Genótipo , Humanos , Masculino , Oseltamivir/administração & dosagem , Oseltamivir/análogos & derivados , Oseltamivir/farmacocinética , Polimorfismo Genético , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética
10.
Clin Pharmacol Ther ; 91(5): 846-55, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22472994

RESUMO

Therapeutic doses of gemfibrozil cause mechanism-based inactivation of CYP2C8 via formation of gemfibrozil 1-O-ß-glucuronide. We investigated the extent of CYP2C8 inactivation caused by three different doses of gemfibrozil twice dailyfor 5 days, using repaglinide as a probe drug, in 10 healthy volunteers. At the end of this 5-day regimen, there were dose-dependent increases in the area under the plasma concentration­time curve from 0 to infinity (AUC0­∞) of repaglinide by3.4-, 5.5-, and 7.0-fold corresponding to 30, 100, and 600 mg of gemfibrozil, respectively, as compared with the control phase (P < 0.001). On the basis of a mechanism-based inactivation model involving gemfibrozil 1-O-ß-glucuronide, a gemfibrozil dose of 30 mg twice daily was estimated to inhibit CYP2C8 by >70% and 100 mg twice daily was estimated to inhibit it by >90%. Hence, gemfibrozil is a strong inactivator of CYP2C8 even in very small, subtherapeutic, multiple doses. Administration of small gemfibrozil doses may be useful in optimizing the pharmacokinetics of CYP2C8 substrate drugs and in reducing the formation of their potentially toxic metabolites via CYP2C8.


Assuntos
Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Genfibrozila/farmacologia , Área Sob a Curva , Hidrocarboneto de Aril Hidroxilases/genética , Glicemia/análise , Carbamatos/metabolismo , Estudos Cross-Over , Citocromo P-450 CYP2C8 , Relação Dose-Resposta a Droga , Genfibrozila/farmacocinética , Genótipo , Glucuronídeos/metabolismo , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado , Transportadores de Ânions Orgânicos/genética , Piperidinas/metabolismo
11.
Br J Pharmacol ; 165(8): 2787-98, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22014153

RESUMO

BACKGROUND AND PURPOSE: Imatinib, a cytochrome P450 2C8 (CYP2C8) and CYP3A4 substrate, markedly increases plasma concentrations of the CYP3A4/5 substrate simvastatin and reduces hepatic CYP3A4/5 activity in humans. Because competitive inhibition of CYP3A4/5 does not explain these in vivo interactions, we investigated the reversible and time-dependent inhibitory effects of imatinib and its main metabolite N-desmethylimatinib on CYP2C8 and CYP3A4/5 in vitro. EXPERIMENTAL APPROACH: Amodiaquine N-deethylation and midazolam 1'-hydroxylation were used as marker reactions for CYP2C8 and CYP3A4/5 activity. Direct, IC(50) -shift, and time-dependent inhibition were assessed with human liver microsomes. KEY RESULTS: Inhibition of CYP3A4 activity by imatinib was pre-incubation time-, concentration- and NADPH-dependent, and the time-dependent inactivation variables K(I) and k(inact) were 14.3 µM and 0.072 in(-1) respectively. In direct inhibition experiments, imatinib and N-desmethylimatinib inhibited amodiaquine N-deethylation with a K(i) of 8.4 and 12.8 µM, respectively, and midazolam 1'-hydroxylation with a K(i) of 23.3 and 18.1 µM respectively. The time-dependent inhibition effect of imatinib was predicted to cause up to 90% inhibition of hepatic CYP3A4 activity with clinically relevant imatinib concentrations, whereas the direct inhibition was predicted to be negligible in vivo. CONCLUSIONS AND IMPLICATIONS: Imatinib is a potent mechanism-based inhibitor of CYP3A4 in vitro and this finding explains the imatinib-simvastatin interaction and suggests that imatinib could markedly increase plasma concentrations of other CYP3A4 substrates. Our results also suggest a possibility of autoinhibition of CYP3A4-mediated imatinib metabolism leading to a less significant role for CYP3A4 in imatinib biotransformation in vivo than previously proposed.


Assuntos
Antineoplásicos/farmacologia , Inibidores do Citocromo P-450 CYP3A , Piperazinas/farmacologia , Pirimidinas/farmacologia , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/metabolismo , Benzamidas , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Humanos , Mesilato de Imatinib , Mucosa Intestinal/metabolismo , Microssomos Hepáticos/enzimologia , Sinvastatina/farmacocinética
12.
Clin Pharmacol Ther ; 89(4): 579-86, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21368757

RESUMO

To study the time to onset of mechanism-based inactivation of cytochrome P450 (CYP) 2C8 by gemfibrozil in vivo, we conducted a randomized five-phase crossover study in 10 healthy volunteers. In one phase the volunteers ingested 0.25 mg of repaglinide alone (control), and in the other phases they received 600 mg of gemfibrozil 0-6 h prior to the repaglinide dose. When gemfibrozil was taken 0, 1, 3, or 6 h before repaglinide, the geometric mean ratio relative to control (90% confidence interval (CI)) of repaglinide area under the plasma concentration-time curve (AUC(0-∞)) was 5.0-fold (4.3-5.7-fold), 6.3-fold (5.4-7.5-fold), 6.6-fold (5.6-7.7-fold), and 5.4-fold (4.8-6.1-fold), respectively (P < 0.001 vs. control). The geometric mean ratio relative to control (90% CI) of the maximum plasma concentration (C(max)) of the CYP2C8-mediated metabolite M4 was 1.0-fold (0.8-1.3-fold), 0.10-fold (0.06-0.17-fold, P < 0.001), 0.06-fold (0.04-0.10-fold, P < 0.001), and 0.09-fold (0.05-0.14-fold, P < 0.001), respectively. The strong inactivation of CYP2C8, evident as soon as 1 h after gemfibrozil dosing, has implications in clinical practice and in studies with gemfibrozil as a CYP2C8 model inhibitor.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Carbamatos/farmacocinética , Genfibrozila/farmacologia , Hipoglicemiantes/farmacocinética , Piperidinas/farmacocinética , Adulto , Área Sob a Curva , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Estudos Cross-Over , Citocromo P-450 CYP2C8 , Feminino , Genfibrozila/análogos & derivados , Genfibrozila/farmacocinética , Glucuronatos/farmacologia , Humanos , Masculino , Fatores de Tempo
13.
Clin Pharmacol Ther ; 88(2): 223-30, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20592724

RESUMO

According to available information, montelukast is metabolized by cytochrome P450 (CYP) 3A4 and 2C9. In order to study the significance of CYP2C8 in the pharmacokinetics of montelukast, 10 healthy subjects were administered gemfibrozil 600 mg or placebo twice daily for 3 days, and 10 mg montelukast on day 3, in a randomized, crossover study. Gemfibrozil increased the mean area under the plasma concentration-time curve (AUC)(0-infinity), peak plasma concentration (C(max)), and elimination half-life (t(1/2)) of montelukast 4.5-fold, 1.5-fold, and 3.0-fold, respectively (P < 0.001). After administration of gemfibrozil, the time to reach C(max) (t(max)) of the montelukast metabolite M6 was prolonged threefold (P = 0.005), its AUC(0-7) was reduced by 40% (P = 0.027), and the AUC(0-24) of the secondary metabolite M4 was reduced by >90% (P < 0.001). In human liver microsomes, gemfibrozil 1-O-beta glucuronide inhibited the formation of M6 (but not of M5) from montelukast 35-fold more potently than did gemfibrozil (half-maximal inhibitory concentration (IC(50)) 3.0 and 107 micromol/l, respectively). In conclusion, gemfibrozil markedly increases the plasma concentrations of montelukast, indicating that CYP2C8 is crucial in the elimination of montelukast.


Assuntos
Acetatos/farmacocinética , Antiasmáticos/farmacocinética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Genfibrozila/efeitos adversos , Hipolipemiantes/efeitos adversos , Antagonistas de Leucotrienos/farmacocinética , Quinolinas/farmacocinética , Acetatos/sangue , Adulto , Antiasmáticos/sangue , Área Sob a Curva , Biotransformação , Estudos Cross-Over , Ciclopropanos , Citocromo P-450 CYP2C8 , DNA/genética , Interações Medicamentosas , Feminino , Genótipo , Glucuronídeos/metabolismo , Meia-Vida , Humanos , Antagonistas de Leucotrienos/sangue , Masculino , Espectrometria de Massas , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Quinolinas/sangue , Sulfetos , Adulto Jovem
14.
Acta Anaesthesiol Scand ; 52(10): 1319-26, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19025521

RESUMO

BACKGROUND: Individual variation in opioid response is considerable, partly due to pharmacokinetic factors. Transporter proteins are becoming increasingly interesting also in the pharmacokinetics of opioids. The efflux transporter P-glycoprotein can affect gastrointestinal absorption and tissue distribution, particularly brain access of many opioids. The aim of this study was to evaluate whether itraconazole, which is a potent inhibitor of P-glycoprotein and CYP3A4, would change the pharmacokinetics or the pharmacodynamics of oral morphine. METHODS: Twelve healthy male volunteers ingested, in a randomized crossover study, once daily 200 mg itraconazole or placebo for 4 days. On day 4, 1 h after the last pre-treatment dose, the subjects ingested 0.3 mg/kg morphine. Blood samples for the determination of plasma morphine, morphine-3-glucuronide (M3G), morphine-6-glucuronide (M6G) and itraconazole concentrations were drawn up to 48 h after morphine ingestion. Pharmacodynamic effects were evaluated using a questionnaire, visual analogue scales, a reaction time test, the Digit Symbol Substitution Test and the Critical Flicker Fusion Test. RESULTS: Itraconazole increased the mean area under the plasma concentration-time curve [AUC (0-9)] of morphine by 29% (P=0.002), its AUC (0-48) by 22% (P=0.013) and its peak plasma concentration by 28% (P=0.035). Itraconazole did not significantly affect the pharmacokinetic variables of M3G or M6G or the pharmacodynamic effects of morphine. CONCLUSIONS: Itraconazole moderately increases plasma concentrations of oral morphine, probably by enhancing its absorption by inhibiting intestinal wall P-glycoprotein. A possible improvement of morphine penetration to the brain could not be observed.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Analgésicos Opioides , Antifúngicos/farmacologia , Itraconazol/farmacologia , Morfina , Administração Oral , Adulto , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/sangue , Área Sob a Curva , Estimulantes do Sistema Nervoso Central/sangue , Estudos Cross-Over , Método Duplo-Cego , Humanos , Masculino , Morfina/administração & dosagem , Morfina/sangue , Derivados da Morfina/sangue , Oxigênio/sangue , Medição da Dor , Projetos de Pesquisa
15.
Clin Pharmacol Ther ; 84(3): 403-11, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18388877

RESUMO

Repaglinide is metabolized by cytochrome P450 (CYP) 2C8 and 3A4. Gemfibrozil has the effect of increasing the area under the concentration-time curve (AUC) of repaglinide eightfold. We studied the effect of dosing interval on the extent of the gemfibrozil-repaglinide interaction. In a randomized five-phase crossover study, 10 healthy volunteers ingested 0.25 mg repaglinide, with or without gemfibrozil pretreatment. Plasma repaglinide, gemfibrozil, their metabolites, and blood glucose were measured. When the last dose of 600 mg gemfibrozil was ingested simultaneously with repaglinide, or 3, 6, or 12 h before, it increased the AUC(0-infinity) of repaglinide 7.0-, 6.5-, 6.2- and 5.0-fold, respectively (P < 0.001). The peak repaglinide concentration increased approximately twofold (P < 0.001), and the half-life was prolonged from 1.2 h to 2-3 h (P < 0.001) during all the gemfibrozil phases. The drug interaction effects persisted at least 12 h after gemfibrozil was administered, although plasma gemfibrozil and gemfibrozil 1-O-beta-glucuronide concentrations were only 5 and 10% of their peak values, respectively. The long-lasting interaction is likely caused by mechanism-based inhibition of CYP2C8 by gemfibrozil glucuronide.


Assuntos
Glicemia/efeitos dos fármacos , Carbamatos/farmacocinética , Genfibrozila/farmacologia , Hipoglicemiantes/farmacocinética , Hipolipemiantes/farmacologia , Piperidinas/farmacocinética , Adulto , Área Sob a Curva , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/genética , Citocromo P-450 CYP2C8 , Interações Medicamentosas , Feminino , Genfibrozila/análogos & derivados , Genfibrozila/sangue , Genfibrozila/farmacocinética , Glucuronatos/sangue , Glucuronatos/farmacocinética , Meia-Vida , Humanos , Hipolipemiantes/sangue , Hipolipemiantes/farmacocinética , Masculino
16.
Clin Pharmacol Ther ; 84(4): 488-96, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19238654

RESUMO

In a randomized crossover study, 24 SLCO181-genotyped healthy volunteers were given daily doses of 1,200 mg gemfibrozil, 40 mg atorvastatin, or placebo, followed by 0.25 mg of repaglinide on day 3. The mean increase in the repaglinide area under the plasma concentration-time curve from 0 h to infinity (AUC(0-infinity)) produced by gemfibrozil was larger in individuals with the SLCO1B1 c.521CC genotype (n = 6) than in those with the c.521TC (n = 6) and c.521TT (n = 12) genotypes, by factors of 1.56 (P = 0.004) and 1.54 (P = 0.002), respectively. Gemfibrozil prolonged the repaglinide elimination half-life 1.43 times more in the c.521 CC group than in the c.521TT group (P = 0.047), but no differences were seen in the effects on peak plasma concentration (C(max)). While on gemfibrozil, the minimum blood glucose concentration after repaglinide intake was 19% lower in the c.521CC participants than in the c.521TT participants (P = 0.009). In the c.521TT group, atorvastatin intake had the effect of increasing repaglinide Cmax and AUC(0-infinity) by41% (P = 0.001) and 18% (P = 0.033), respectively. In conclusion, the extent of gemfibrozil-repaglinide interaction depends on SLCO1B1 genotype. Atorvastatin raises plasma repaglinide concentrations, probably by inhibiting organic anion transporting polypeptide 1B1 (OATP1B1).


Assuntos
Carbamatos/farmacocinética , Genfibrozila/farmacocinética , Ácidos Heptanoicos/farmacocinética , Hipolipemiantes/farmacocinética , Transportadores de Ânions Orgânicos/genética , Piperidinas/farmacocinética , Polimorfismo Genético , Pirróis/farmacocinética , Administração Oral , Adulto , Área Sob a Curva , Atorvastatina , Glicemia/análise , Carbamatos/administração & dosagem , Carbamatos/sangue , Estudos Cross-Over , Interações Medicamentosas , Feminino , Genfibrozila/administração & dosagem , Genfibrozila/sangue , Genótipo , Meia-Vida , Ácidos Heptanoicos/administração & dosagem , Ácidos Heptanoicos/sangue , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/sangue , Hipoglicemiantes/farmacocinética , Hipolipemiantes/administração & dosagem , Hipolipemiantes/sangue , Transportador 1 de Ânion Orgânico Específico do Fígado , Masculino , Transportadores de Ânions Orgânicos/metabolismo , Piperidinas/administração & dosagem , Piperidinas/sangue , Pirróis/administração & dosagem , Pirróis/sangue
17.
Pharmacogenomics J ; 8(4): 268-77, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17923851

RESUMO

Cytochrome P450 2C8 (CYP2C8) plays a major role in the metabolism of therapeutically important drugs which exhibit large interindividual differences in their pharmacokinetics. In order to evaluate any genetic influence on this variation, a CYP2C8 phenotype-genotype evaluation was carried out in Caucasians. Two novel CYP2C8 haplotypes, named B and C with frequencies of 24 and 22% in Caucasians, respectively, were identified and caused a significantly increased and reduced paclitaxel 6alpha-hydroxylation, respectively, as evident from analyses of 49 human liver samples. In healthy white subjects, CYP2C8*3 and the two novel haplotypes significantly influenced repaglinide pharmacokinetics in SLCO1B1c.521T/C heterozygous individuals: haplotype B was associated with reduced and haplotype C with increased repaglinide AUC (0-infinity). Functional studies suggested -271C>A (CYP2C8*1B) as a causative SNP in haplotype B. In conclusion, two novel common CYP2C8 haplotypes were identified and significantly associated with altered rate of CYP2C8-dependent drug metabolism in vitro and in vivo.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Carbamatos/metabolismo , Haplótipos/genética , Paclitaxel/metabolismo , Piperidinas/metabolismo , Hidrocarboneto de Aril Hidroxilases/metabolismo , Hidrocarboneto de Aril Hidroxilases/fisiologia , Carbamatos/farmacologia , Citocromo P-450 CYP2C8 , Variação Genética/efeitos dos fármacos , Variação Genética/genética , Haplótipos/efeitos dos fármacos , Humanos , Paclitaxel/farmacologia , Piperidinas/farmacologia , População Branca/genética
18.
Br J Clin Pharmacol ; 64(6): 772-84, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17662086

RESUMO

AIMS: To use population pharmacokinetic modelling to characterize the influence of developmental and demographic factors on the pharmacokinetic variability of ciclosporin. METHODS: Pharmacokinetic modelling was performed in NONMEM using a dataset comprising 162 pretransplant children, aged 0.36-17.5 years. Ciclosporin was given intravenously (3 mg kg(-1)) and orally (10 mg kg(-1)) on separate occasions followed by blood sampling for 24 h. RESULTS: A three-compartment model with first-order absorption without lag-time best described the pharmacokinetics of ciclosporin. The most important covariate affecting systemic clearance (CL) and distribution volume (V) was body weight (BW; scaled allometrically), responsible for a fourfold difference in uncorrected ciclosporin CL and a sixfold difference in ciclosporin V. The other significant covariates, haematocrit, plasma cholesterol and creatinine, were estimated to explain 20-30% of interindividual differences in CL and V of ciclosporin. No age-related changes in oral bioavailability or in BW-normalized V were seen. The BW-normalized CL (CL/BW) declined with age and prepubertal children (<8 years) had an approximately 25% higher CL/BW than did older children. Normalization of CL for allometric BW (BW(3/4)) removed its relationship to age. CONCLUSION: The relationship between CL and allometric BW is consistent with a gradual reduction in relative liver size, until adult values, and a relatively constant CYP3A4 content in the liver from about 6-12 months of age to adulthood. Ciclosporin oral bioavailability, known previously to display large interindividual variability, is not influenced by age. These findings can enable better individualization of ciclosporin dosing in infants, children and adolescents.


Assuntos
Ciclosporina/farmacocinética , Transplante de Rim , Modelos Biológicos , Adolescente , Fatores Etários , Criança , Pré-Escolar , Bases de Dados Factuais , Humanos , Lactente , Taxa de Depuração Metabólica/efeitos dos fármacos , Taxa de Depuração Metabólica/fisiologia
19.
Clin Pharmacol Ther ; 81(6): 833-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17392729

RESUMO

Case reports suggest that cranberry juice can increase the anticoagulant effect of warfarin. We investigated the effects of cranberry juice on R-S-warfarin, tizanidine, and midazolam; probes of CYP2C9, CYP1A2, and CYP3A4. Ten healthy volunteers took 200 ml cranberry juice or water t.i.d. for 10 days. On day 5, they ingested 10 mg racemic R-S-warfarin, 1 mg tizanidine, and 0.5 mg midazolam, with juice or water, followed by monitoring of drug concentrations and thromboplastin time. Cranberry juice did not increase the peak plasma concentration or area under concentration-time curve (AUC) of the probe drugs or their metabolites, but slightly decreased (7%; P=0.051) the AUC of S-warfarin. Cranberry juice did not change the anticoagulant effect of warfarin. Daily ingestion of cranberry juice does not inhibit the activities of CYP2C9, CYP1A2, or CYP3A4. A pharmacokinetic mechanism for the cranberry juice-warfarin interaction seems unlikely.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Bebidas , Clonidina/análogos & derivados , Citocromo P-450 CYP1A2/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Alimento-Droga , Midazolam/farmacocinética , Vaccinium macrocarpon , Varfarina/farmacocinética , Adulto , Anticoagulantes/química , Anticoagulantes/farmacocinética , Área Sob a Curva , Clonidina/farmacocinética , Estudos Cross-Over , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP3A , Feminino , Meia-Vida , Humanos , Masculino , Estereoisomerismo , Varfarina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA