Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Transl Sci ; 16(6): 1039-1048, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37085998

RESUMO

Letermovir is approved for use in cytomegalovirus-seropositive hematopoietic stem cell transplant recipients and is investigated in other transplant settings. Nonlinear pharmacokinetics (PKs) were observed in clinical studies after intravenous and oral dosing across a wide dose range, including the efficacious doses of 240 and 480 mg. A physiologically-based PK (PBPK) model for letermovir was built to develop a plausible explanation for the nonlinear PKs observed in clinical studies. In vitro studies suggested that letermovir elimination and distribution are mediated by saturable uridine glucuronosyltransferases (UGT)-metabolism and by saturable hepatic uptake via organic anion-transporting polypeptides (OATP) 1B. A sensitivity analysis of parameters describing the metabolism and distribution mechanisms indicated that the greater than dose-proportional increase in letermovir exposure is best described by a saturable OATP1B-mediated transport. This PBPK model was further used to evaluate the drug interaction potential between letermovir and everolimus, an immunosuppressant that may be co-administered with letermovir depending on regions. Because letermovir inhibits cytochrome P450 (CYP) 3A and everolimus is a known CYP3A substrate, an interaction when concomitantly administered is anticipated. The drug-drug interaction simulation confirmed that letermovir will likely increase everolimus are under the curve by 2.5-fold, consistent with the moderate increase in exposure observed with midazolam in the clinic. The output highlights the importance of drug monitoring, which is common clinical practice for everolimus to maintain safe and efficacious drug concentrations in the targeted patient population when concomitantly administered with letermovir.


Assuntos
Everolimo , Imunossupressores , Humanos , Everolimo/efeitos adversos , Interações Medicamentosas , Imunossupressores/farmacocinética , Acetatos , Citocromo P-450 CYP3A/metabolismo , Modelos Biológicos
2.
Eur J Drug Metab Pharmacokinet ; 42(2): 319-326, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27101423

RESUMO

BACKGROUND AND OBJECTIVES: Prediction of metabolic clearance has been a challenge for compounds exhibiting minimal turnover in typical in vitro stability experiments. The aim of the current study is to evaluate the utilization of plated human hepatocytes to predict intrinsic clearance of low-turnover compounds. METHODS: The disappearance of test compounds was determined for up to 48 h while enzyme activities in plated hepatocytes were monitored concurrently in a complimentary experiment. RESULTS: Consistent with literature reports, marked time-dependent loss of cytochrome P450 (CYP) enzyme activities was observed during the 48-h incubation period. To account for the loss of enzyme activities, a term "fraction of activity remaining" was calculated based on area-under-the-curve derived from the average rate of activity loss (k avg), and then applied as a correction factor for intrinsic clearance determination. Twelve compounds were selected in this study to cover phase I and phase II biotransformation pathways, with in vivo intrinsic clearance values, representing metabolic clearance only, ranging from 0.66 to 47 ml/min/kg. Determination of in vitro intrinsic clearance using three individual preparations of hepatocytes revealed a reasonably good agreement (generally within threefold) between the predicted and the observed metabolic clearance for all 12 compounds tested. CONCLUSIONS: The current results indicated that plated hepatocytes can be utilized to provide clearance predictions for compounds with low-turnover in humans when corrected for the loss in enzyme activities.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Hepatócitos/metabolismo , Preparações Farmacêuticas/metabolismo , Células Cultivadas , Hepatócitos/enzimologia , Humanos , Taxa de Depuração Metabólica , Fatores de Tempo
3.
Drug Metab Dispos ; 41(3): 668-81, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23293300

RESUMO

The inhibitory effect of boceprevir (BOC), an inhibitor of hepatitis C virus nonstructural protein 3 protease was evaluated in vitro against a panel of drug-metabolizing enzymes and transporters. BOC, a known substrate for cytochrome P450 (P450) CYP3A and aldo-ketoreductases, was a reversible time-dependent inhibitor (k(inact) = 0.12 minute(-1), K(I) = 6.1 µM) of CYP3A4/5 but not an inhibitor of other major P450s, nor of UDP-glucuronosyltransferases 1A1 and 2B7. BOC showed weak to no inhibition of breast cancer resistance protein (BCRP), P-glycoprotein (Pgp), or multidrug resistance protein 2. It was a moderate inhibitor of organic anion transporting polypeptide (OATP) 1B1 and 1B3, with an IC(50) of 18 and 4.9 µM, respectively. In human hepatocytes, BOC inhibited CYP3A-mediated metabolism of midazolam, OATP1B-mediated hepatic uptake of pitavastatin, and both the uptake and metabolism of atorvastatin. The inhibitory potency of BOC was lower than known inhibitors of CYP3A (ketoconazole), OATP1B (rifampin), or both (telaprevir). BOC was a substrate for Pgp and BCRP but not for OATP1B1, OATP1B3, OATP2B1, organic cation transporter, or sodium/taurocholate cotransporting peptide. Overall, our data suggest that BOC has the potential to cause pharmacokinetic interactions via inhibition of CYP3A and CYP3A/OATP1B interplay, with the interaction magnitude lower than those observed with known potent inhibitors. Conversely, pharmacokinetic interactions of BOC, either as a perpetrator or victim, via other major P450s and transporters tested are less likely to be of clinical significance. The results from clinical drug-drug interaction studies conducted thus far are generally supportive of these conclusions.


Assuntos
Antivirais/metabolismo , Inibidores Enzimáticos/metabolismo , Enzimas/metabolismo , Fígado/enzimologia , Moduladores de Transporte de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Prolina/análogos & derivados , Animais , Antivirais/toxicidade , Biotransformação , Células CHO , Cricetinae , Cricetulus , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A , Cães , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/toxicidade , Enzimas/genética , Feminino , Glucuronosiltransferase/metabolismo , Humanos , Cinética , Células LLC-PK1 , Fígado/efeitos dos fármacos , Transportador 1 de Ânion Orgânico Específico do Fígado , Células Madin Darby de Rim Canino , Masculino , Moduladores de Transporte de Membrana/toxicidade , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Microssomos Hepáticos/enzimologia , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/metabolismo , Oxirredutases/metabolismo , Prolina/metabolismo , Prolina/toxicidade , Proteínas Recombinantes/metabolismo , Suínos , Transfecção
4.
J Pharmacol Exp Ther ; 325(3): 935-46, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18310472

RESUMO

After oral treatment (once daily) for 4 weeks with the potent bradykinin B(1) receptor antagonist methyl 3-chloro-3'-fluoro-4'-{(1R)-1-[({1-[(trifluoroacetyl)amino]cyclopropyl}carbonyl)-amino]ethyl}-1,1'-biphenyl-2-carboxylate (MK-0686), rhesus monkeys (Macaca mulatta) exhibited significantly reduced systemic exposure of the compound in a dose-dependent manner, suggesting an occurrence of autoinduction of MK-0686 metabolism. This possibility is supported by two observations. 1) MK-0686 was primarily eliminated via biotransformation in rhesus monkeys, with oxidation on the chlorophenyl ring as one of the major metabolic pathways. This reaction led to appreciable formation of a dihydrodiol (M11) and a hydroxyl (M13) product in rhesus liver microsomes supplemented with NADPH. 2) The formation rate of these two metabolites determined in liver microsomes from MK-0686-treated groups was > or = 2-fold greater than the value for a control group. Studies with recombinant rhesus P450s and monoclonal antibodies against human P450 enzymes suggested that CYP2C75 played an important role in the formation of M11 and M13. The induction of this enzyme by MK-0686 was further confirmed by a concentration-dependent increase of its mRNA in rhesus hepatocytes, and, more convincingly, the enhanced CYP2C proteins and catalytic activities toward CYP2C75 probe substrates in liver microsomes from MK-0686-treated animals. Furthermore, a good correlation was observed between the rates of M11 and M13 formation and hydroxylase activities toward probe substrates determined in a panel of liver microsomal preparations from control and MK-0686-treated animals. Therefore, MK-0686, both a substrate and inducer for CYP2C75, caused autoinduction of its own metabolism in rhesus monkeys by increasing the expression of this enzyme.


Assuntos
Acetamidas/farmacocinética , Benzoatos/farmacocinética , Antagonistas de Receptor B1 da Bradicinina , Sistema Enzimático do Citocromo P-450/metabolismo , Acetamidas/sangue , Acetamidas/urina , Animais , Benzoatos/sangue , Benzoatos/urina , Bile/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Sistema Enzimático do Citocromo P-450/genética , Feminino , Hepatócitos/metabolismo , Humanos , Macaca mulatta , Masculino , Microssomos Hepáticos/metabolismo , Receptor de Pregnano X , Receptor B1 da Bradicinina/metabolismo , Receptores de Esteroides/metabolismo , Proteínas Recombinantes/metabolismo
5.
Pharm Res ; 22(10): 1658-66, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16180123

RESUMO

PURPOSE: Phenethyl isothiocyanate (PEITC) is a dietary component present in cruciferous vegetables and reported to have chemopreventive properties. Previous reports of PEITC pharmacokinetics have measured total ITC (PEITC and its metabolites) in plasma. Our objective was to examine the dose-dependent pharmacokinetics and oral bioavailability of unchanged PEITC, as well as its pH- and temperature-dependent stability and its serum protein binding. METHODS: Stability was studied at different pH values at room temperature and 4 degrees C. Protein binding was determined by equilibrium dialysis. For the pharmacokinetics study, male Sprague-Dawley rats were administered with PEITC at doses of 2, 10, 100, or 400 micromol/kg intravenously or 10 or 100 micromol/kg orally. Plasma samples were analyzed by liquid chromatography-tandem mass spectrometry. Pharmacokinetic analysis was conducted by WinNonlin and ADAPT II. RESULTS: Phenethyl isothiocyanate was stable in aqueous buffers at pH 7.4 with half-lives of 56.1 and 108 h at room temperature and 4 degrees C, respectively. The free fraction of PEITC in rat serum was 0.019. The clearance (Cl) at a low dose of PEITC (2 micromol/kg) was 0.70 +/- 0.17 L h(-1) kg(-1) with an apparent volume of distribution (Vss) of 1.94 +/- 0.42 L/kg. At higher doses, Cl tended to decrease, whereas Vss increased. Oral bioavailability of PEITC was 115 and 93% at doses of 10 and 100 micromol/kg, respectively. A three-compartment model with Michaelis-Menten elimination and distribution was found to best characterize the plasma concentration profiles. CONCLUSIONS: Phenethyl isothiocyanate is stable in biological samples, with increased stability under refrigerated conditions. It has high oral bioavailability, low clearance, and high protein binding in rats; nonlinear elimination and distribution occur following the administration of high doses. This investigation represents the first report of the pharmacokinetics of dietary PEITC.


Assuntos
Isotiocianatos/farmacologia , Isotiocianatos/farmacocinética , Administração Oral , Animais , Anticarcinógenos/farmacocinética , Anticarcinógenos/farmacologia , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Concentração de Íons de Hidrogênio , Injeções Intravenosas , Isotiocianatos/metabolismo , Masculino , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Albumina Sérica/metabolismo , Fatores de Tempo
6.
Chem Res Toxicol ; 18(6): 934-45, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15962928

RESUMO

The 2,3-diaminopyridine (DAP) moiety was found to represent a core structure essential for the potency of a new series of human bradykinin B(1) receptor antagonists. However, incubation of (14)C-labeled 2,3-DAP derivatives with rat and human liver microsomes resulted in substantial irreversible binding of radioactive material to macromolecules by a process that was NADPH-dependent. Trapping the reactive species with GSH led to significant reduction of the irreversible binding of radioactivity, with concomitant formation of abundant GSH adducts. One type of thiol adducts (detected in both human and rat liver microsomes), resulting from addition of 305 Da to the parent compound, was observed with all 2,3-DAP compounds. These adducts also were detected in rat hepatocyte incubates, as well as in rat bile, following intravenous administration of 2,3-DAPs. Formation of the conjugates appeared to involve modification of the DAP ring, based upon mass spectral analysis of a number of representative GSH adducts; this was corroborated by detailed LC NMR analysis of one compound. Formation of this type of GSH conjugate was markedly reduced when the 2-amino methyl group linking the 2,3-DAP and the biphenyl moiety was replaced with an ether oxygen atom. It is postulated, therefore, that oxidation of the 2-amino group serves as a key step leading to the formation of reactive species associated with the DAP core. In addition, this step appears to be mediated primarily by P450 3A, as evidenced by the marked decrease in both the irreversible binding of radioactivity and the formation of the GSH adducts in human liver microsomes following treatment with ketoconazole and monoclonal antibodies against P450 3A. A mechanism for the bioactivation of 2,3-DAP is proposed wherein oxidation (dehydrogenation or N-hydroxylation followed by dehydration) of the 2-amino group, catalyzed by P450 3A, results in the formation of a highly electrophilic species, pyridine-2,3-diimine.


Assuntos
Aminopiridinas/metabolismo , Antagonistas de Receptor B1 da Bradicinina , Glutationa/metabolismo , Microssomos Hepáticos/metabolismo , Aminopiridinas/farmacocinética , Animais , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Oxirredução , Ligação Proteica , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA