Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biopharm Drug Dispos ; 40(2): 62-69, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30652318

RESUMEN

The hepatic uptake of clinical drugs mediated by human hepatic organic anion transporting polypeptides (OATP/SLCO) has been reported extensively. In this study, hepatic uptake by recombinantly expressed monkey OATP1B1, OATP1B3 and OATP2B1 was investigated using three human OATP1B1 and OATP1B3 substrates (pitavastatin, pravastatin and rosuvastatin) and one OATP1B3 substrate (telmisartan), as the governmental drug interaction guidelines recommend, and seven reported clinical drugs. The uptake of known human probes into recombinant OATP-expressing cells was significantly greater than that of mock cells. Consequently, pitavastatin, pravastatin and rosuvastatin were suggested to be substrates of recombinant monkey OATP1B1 and OATP1B3, and telmisartan was suggested to be a substrate of recombinant monkey OATP1B3, in a manner similar to human OATPs. In contrast, atorvastatin, bosentan, etoposide, fexofenadine, fluvastatin, glibenclamide and simeprevir were broadly transported by recombinant monkey OATP1B1, OATP1B3 and OATP2B1. Furthermore, some of the 16 non-synonymous monkey OATP1B1 variants found in 64 cynomolgus and 32 rhesus monkeys mediated up to a 1.6-fold [3 H]pitavastatin uptake (with low Michaelis constant values) in comparison with the wild type under the present conditions. Despite sequences of monkey recombinant OATPs not being totally reflective of those of human OATPs, our results collectively suggested that OATP1B1, OATP1B3 or OATP2B1 in monkeys could mediate roughly a similar hepatic uptake of various OATP probes. Recombinant monkey OATPs would be good experimental tools for in vitro hepatic uptake in cell systems.


Asunto(s)
Transportadores de Anión Orgánico/metabolismo , Quinolinas/metabolismo , Proteínas Recombinantes/metabolismo , Telmisartán/metabolismo , Animales , Interacciones Farmacológicas , Células HEK293 , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado/genética , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Macaca fascicularis , Macaca mulatta , Transportadores de Anión Orgánico/genética , Plásmidos , Pravastatina/metabolismo , Proteínas Recombinantes/genética , Rosuvastatina Cálcica/metabolismo , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/genética , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/metabolismo , Especificidad por Sustrato
2.
Drug Metab Dispos ; 45(9): 1027-1034, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28698303

RESUMEN

UDP-Glucuronosyltransferases (UGTs) are major phase II drug-metabolizing enzymes. Each member of the UGT family exhibits a unique but occasionally overlapping substrate specificity and tissue-specific expression pattern. Earlier studies have reported that human UGT1A10 is expressed in the gastrointestinal tract at the mRNA level, but the evaluation at the protein level, especially tissue or cellular localization, has lagged behind because of the lack of a specific antibody. In this study, we prepared a monoclonal antibody to UGT1A10 to elucidate the tissue/cellular distribution and interindividual variability of UGT1A10 protein expression. Western blot analysis revealed that the prepared antibody does not cross-react with any other human UGTs. Using this specific antibody, we observed that UGT1A10 protein is expressed in the small intestine but not in the liver or kidney. Immunohistochemical analysis revealed the expression of UGT1A10 protein in epithelial cells of the crypts and villi of the duodenum. In the small intestine microsomes from six individuals, the UGT1A10 protein levels exhibited 16-fold variability. Dopamine 3- and 4-glucuronidation, which is mainly catalyzed by UGT1A10 and by other UGT isoforms marginally, exhibited 50- to 65-fold variability, and they were not correlated with the UGT1A10 protein levels. Interestingly, the enzymatic activities of recombinant UGT1A10 in insect cells that were normalized to the UGT1A10 protein level were markedly lower than those in pooled human small intestine microsomes. Thus, the UGT1A10 antibody we generated made it possible to investigate the tissue/cellular distribution and interindividual variability of UGT1A10 protein expression for understanding the pharmacological and toxicological role of UGT1A10.


Asunto(s)
Anticuerpos Monoclonales/química , Glucuronosiltransferasa/metabolismo , Intestinos/enzimología , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Células Epiteliales/citología , Células Epiteliales/enzimología , Femenino , Glucuronosiltransferasa/biosíntesis , Glucuronosiltransferasa/inmunología , Células HEK293 , Células Hep G2 , Humanos , Intestinos/citología , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley
3.
Drug Metab Dispos ; 44(7): 888-96, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27112166

RESUMEN

Glucuronidation, an important phase II metabolic route, is generally considered to be a detoxification pathway. However, acyl glucuronides (AGs) have been implicated in the toxicity of carboxylic acid drugs due to their electrophilic reactivity. Zomepirac (ZP) was withdrawn from the market because of adverse effects such as renal toxicity. Although ZP is mainly metabolized to acyl glucuronide (ZP-AG) by UDP-glucuronosyltransferase, the role of ZP-AG in renal toxicity is unknown. In this study, we established a ZP-induced kidney injury mouse model by pretreatment with tri-o-tolyl phosphate (TOTP), a nonselective esterase inhibitor, and l-buthionine-(S,R)-sulfoximine (BSO), a glutathione synthesis inhibitor. The role of ZP-AG in renal toxicity was investigated using this model. The model showed significant increases in blood urea nitrogen (BUN) and creatinine (CRE), but not alanine aminotransferase. The ZP-AG concentrations were elevated by cotreatment with TOTP in the plasma and liver and especially in the kidney. The ZP-AG concentrations in the kidney correlated with values for BUN and CRE. Upon histopathological examination, vacuoles and infiltration of mononuclear cells were observed in the model mouse. In addition to immune-related responses, oxidative stress markers, such as the glutathione/disulfide glutathione ratio and malondialdehyde levels, were different in the mouse model. The suppression of ZP-induced kidney injury by tempol, an antioxidant agent, suggested the involvement of oxidative stress in ZP-induced kidney injury. This is the first study to demonstrate that AG accumulation in the kidney by TOTP and BSO treatment could explain renal toxicity and to show the in vivo toxicological potential of AGs.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Riñón/efectos de los fármacos , Tolmetina/análogos & derivados , Lesión Renal Aguda/sangre , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Antioxidantes/farmacología , Biomarcadores/sangre , Biotransformación , Nitrógeno de la Urea Sanguínea , Butionina Sulfoximina/farmacología , Creatinina/sangre , Óxidos N-Cíclicos/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Femenino , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Marcadores de Spin , Factores de Tiempo , Tolmetina/administración & dosificación , Tolmetina/sangre , Tolmetina/toxicidad , Tritolilfosfatos/farmacología
4.
Biopharm Drug Dispos ; 37(3): 142-155, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26749565

RESUMEN

We investigated the tissue distribution of teneligliptin, a dipeptidyl peptidase (DPP)-4 inhibitor, in rats, and compared it with tissue distributions previously reported for other DPP-4 inhibitors. Following the oral administration of [14 C]teneligliptin to Sprague-Dawley rats, it was predominantly distributed to the kidney and liver, followed by the lung, spleen, and pituitary gland. The elimination half-life (t1/2 ) of [14 C]teneligliptin was 68.3 and 69.0 h in the kidney and liver, respectively; these values were about 10 times greater than the plasma t1/2 . Of note, the elimination of [14 C]teneligliptin from tissues with high DPP-4 activity (kidney, liver, and lung) was slower in wild-type rats than in DPP-4-deficient rats, especially in the kidney. By contrast, in the heart and pancreas, which weakly express DPP-4, we observed no difference in [14 C]teneligliptin concentrations between the two animal strains. In the kidney, most radioactivity was attributable to unchanged teneligliptin from 0.5 to 72 h after administration. The marked difference in the distribution of [14 C]teneligliptin between the two strains suggests that the high binding affinity of teneligliptin for DPP-4 is involved in its tissue distribution. The currently marketed DPP-4 inhibitors are highly distributed to the liver, kidney, and lung, but the extent of tissue distribution varies greatly among the drugs. The differences in the tissue distributions of DPP-4 inhibitors might be related to differences in their pleiotropic effects. This article is protected by copyright. All rights reserved.

5.
Biopharm Drug Dispos ; 37(8): 479-490, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27603548

RESUMEN

Cyclosporine A, an inhibitor of hepatic organic anion transporting polypeptides (OATPs), reportedly increased plasma concentrations of probe substrates, although its maximum unbound blood concentrations were lower than the experimental half-maximal inhibitory (IC50 ) concentrations. Pre-incubation with cyclosporine A in vitro before simultaneous incubation with probes has been reported to potentiate its inhibitory effects on recombinant human OATP-mediated probe uptake. In the present study, the effects of cyclosporine A and rifampicin on recombinant cynomolgus monkey OATP-mediated pitavastatin uptake were investigated in pre- and simultaneous incubation systems. Pre-incubation with cyclosporine A, but not with rifampicin, decreased the apparent IC50 values on recombinant cynomolgus monkey OATP1B1- and OATP1B3-mediated pitavastatin uptake. Application of the co-incubated IC50 values toward R values (1 + [unbound inhibitor]inlet to the liver, theoretically maximum /inhibition constant) in static models, 1.1 in monkeys and 1.3 in humans, for recombinant cynomolgus monkey and human OATP1B1-mediated pitavastatin uptake might result in the poor prediction of drug interaction magnitudes. In contrast, the lowered IC50 values after pre-incubation with cyclosporine A provided better prediction with R values of 3.9 for monkeys and 2.7 for humans when the estimated maximum cyclosporine A concentrations at the inlet to the liver were used. These results suggest that the enhanced inhibitory potential of perpetrator medicines by pre-incubation on cynomolgus monkey OATP-mediated pitavastatin uptake in vitro could be of value for the precise estimation of drug interaction magnitudes in silico, in accordance with the findings from pre-administration of inhibitors on pitavastatin pharmacokinetics validated in monkeys. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Antifúngicos/metabolismo , Ciclosporina/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado/biosíntesis , Transportadores de Anión Orgánico Sodio-Independiente/biosíntesis , Quinolinas/metabolismo , Animales , Antifúngicos/farmacología , Transporte Biológico/fisiología , Ciclosporina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Células HEK293 , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Macaca fascicularis , Quinolinas/farmacología , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos
6.
Xenobiotica ; 45(6): 556-62, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25539456

RESUMEN

1. Raloxifene-6-glucuronide (R6G) is a substrate of rat multidrug resistance-associated protein 2 (Mrp2), a transporter responsible for biliary excretion of organic anions. 2. Pharmacokinetic modeling of R6G in Eisai hyperbilirubinemic rats (EHBRs), hereditary Mrp2-deficient rats, and wild-type Sprague-Dawley rats (SDRs) indicated that reduction in not only biliary excretion but also hepatic uptake of R6G influenced low clearance in EHBRs. 3. An integration plot study demonstrated that the hepatic uptake of R6G was 66% lower in EHBRs than that in SDRs. A reduction was observed for the other Mrp2 substrate Valsartan (95% lower) but not for estradiol-17ß-glucuronide (E217ßG). This variation may be associated with the difference in substrate specificity of transporters and/or inhibition of hepatic uptake of organic anions by endogenous substances such as bilirubin glucuronides. 4. In conclusion, incidental alteration of the hepatic uptake of organic anions should be considered as an explanation of their enhanced systemic exposure in EHBRs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Glucuronatos/farmacocinética , Hígado/metabolismo , Piperidinas/farmacocinética , Valsartán/farmacocinética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Estradiol/análogos & derivados , Estradiol/farmacocinética , Estradiol/farmacología , Glucuronatos/farmacología , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/metabolismo , Masculino , Piperidinas/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Valsartán/farmacología
7.
Xenobiotica ; 44(3): 242-53, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23855261

RESUMEN

1. The absorption, metabolism and excretion of teneligliptin were investigated in healthy male subjects after a single oral dose of 20 mg [(14)C]teneligliptin. 2. Total plasma radioactivity reached the peak concentration at 1.33 h after administration and thereafter disappeared in a biphasic manner. By 216 h after administration, ≥90% of the administered radioactivity was excreted, and the cumulative excretion in the urine and faeces was 45.4% and 46.5%, respectively. 3. The most abundant metabolite in plasma was a thiazolidine-1-oxide derivative (designated as M1), which accounted for 14.7% of the plasma AUC (area under the plasma concentration versus time curve) of the total radioactivity. The major components excreted in urine were teneligliptin and M1, accounting for 14.8% and 17.7% of the dose, respectively, by 120 h, whereas in faeces, teneligliptin was the major component (26.1% of the dose), followed by M1 (4.0%). 4. CYP3A4 and FMO3 are the major enzymes responsible for the metabolism of teneligliptin in humans. 5. This study indicates the involvement of renal excretion and multiple metabolic pathways in the elimination of teneligliptin from the human body. Teneligliptin is unlikely to cause conspicuous drug interactions or changes in its pharmacokinetics patients with renal or hepatic impairment, due to a balance in the elimination pathways.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacocinética , Oxigenasas/metabolismo , Pirazoles/metabolismo , Pirazoles/farmacocinética , Tiazolidinas/metabolismo , Tiazolidinas/farmacocinética , Absorción , Adulto , Área Bajo la Curva , Radioisótopos de Carbono/sangre , Radioisótopos de Carbono/farmacocinética , Cromatografía Líquida de Alta Presión , Inhibidores de la Dipeptidil-Peptidasa IV/sangre , Inhibidores de la Dipeptidil-Peptidasa IV/química , Heces/química , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Modelos Biológicos , Estructura Molecular , Pirazoles/sangre , Pirazoles/química , Conteo por Cintilación , Tiazolidinas/sangre , Tiazolidinas/química , Factores de Tiempo
8.
Drug Metab Dispos ; 41(10): 1875-82, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23929936

RESUMEN

Drug-drug interactions (DDIs) caused by the inhibition of hepatic uptake transporters such as organic anion transporting polypeptide (OATP) can affect therapeutic efficacy and cause adverse reactions. We investigated the potential utility of pitavastatin as an in vivo probe substrate for preclinically studying OATP-mediated DDIs using cynomolgus monkeys. Cyclosporine A (CsA) and rifampicin (RIF), typical OATP inhibitors, inhibited active uptake of pitavastatin into monkey hepatocytes with half-maximal inhibitory concentration values comparable with those in human hepatocytes. CsA and RIF increased the area under the plasma concentration-time curve (AUC) of intravenously administered pitavastatin in cynomolgus monkeys by 3.2- and 3.6-fold, respectively. In addition, there was no apparent prolongation of the elimination half-life of pitavastatin due to the decrease in both hepatic clearance and volume of distribution. These findings suggest that DDIs were caused by the inhibition of hepatic uptake of pitavastatin. CsA and RIF increased the AUC of orally administered pitavastatin by 10.6- and 14.8-fold, respectively, which was additionally caused by the effect of the CsA and RIF in the gastrointestinal tract. Hepatic contribution to the overall DDI for oral pitavastatin with CsA was calculated from the changes in hepatic availability and clearance, and it was shown that the magnitude of hepatic DDI was comparable between the present study and the clinical study. In conclusion, pharmacokinetic studies using pitavastatin as a probe in combination with drug candidates in cynomolgus monkeys are useful to support the assessment of potential clinical DDIs involving hepatic uptake transporters.


Asunto(s)
Interacciones Farmacológicas/genética , Hígado/metabolismo , Macaca fascicularis/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Quinolinas/farmacología , Quinolinas/farmacocinética , Animales , Área Bajo la Curva , Transporte Biológico/genética , Tracto Gastrointestinal/metabolismo , Semivida , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/efectos de los fármacos , Macaca fascicularis/genética , Masculino , ARN Mensajero/genética
9.
Chem Commun (Camb) ; 59(34): 5071-5074, 2023 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37021390

RESUMEN

We report that a selective fluorescent indicator NBD-NCD for UGGAA repeats resulted in fluorescence quenching upon binding to RNA and recovered the fluorescence by displacing NBD-NCD with UGGAA repeat-targeted small molecules. The fluorescent indicator displacement assay using NBD-NCD can detect the interaction of small molecules with UGGAA repeats.


Asunto(s)
Enfermedades no Transmisibles , Humanos , Colorantes Fluorescentes/química , ARN/química , Espectrometría de Fluorescencia
10.
Drug Metab Dispos ; 39(9): 1495-502, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21646435

RESUMEN

Raloxifene is extensively glucuronidated in humans, effectively reducing its oral bioavailability (2%). It was also reported to be glucuronidated in preclinical animals, but its effects on the oral bioavailability have not been fully elucidated. In the present study, raloxifene and its glucuronides in the portal and systemic blood were monitored in Gunn rats deficient in UDP-glucuronosyltransferase (UGT) 1A, Eisai hyperbilirubinemic rats (EHBRs), which hereditarily lack multidrug resistance-associated protein (MRP) 2, and wild-type rats after oral administration. The in vitro-in vivo correlation (IVIVC) of four UGT substrates (raloxifene, biochanin A, gemfibrozil, and mycophenolic acid) in rats was also evaluated. In Gunn rats, the product of fraction absorbed and intestinal availability and hepatic availability of raloxifene were 0.63 and 0.43, respectively; these values were twice those observed in wild-type Wistar rats, indicating that raloxifene was glucuronidated in both the liver and intestine. The ratio of glucuronides to unchanged drug in systemic blood was substantially higher in EHBRs (129-fold) than in the wild-type Sprague-Dawley rats (10-fold), suggesting the excretion of raloxifene glucuronides caused by MRP2. The IVIVC of the other UGT substrates in rats displayed a good relationship, but the oral clearance values of raloxifene and biochanin A, which were extensively glucuronidated by rat intestinal microsomes, were higher than the predicted clearances using rat liver microsomes, suggesting that intestinal metabolism may be a great contributor to the first-pass effect. Therefore, evaluation of intestinal and hepatic glucuronidation for new chemical entities is important to improve their pharmacokinetic profiles.


Asunto(s)
Glucurónidos/metabolismo , Mucosa Intestinal/metabolismo , Clorhidrato de Raloxifeno/farmacocinética , Animales , Disponibilidad Biológica , Perros , Descubrimiento de Drogas/métodos , Glucuronosiltransferasa/metabolismo , Hígado/metabolismo , Macaca fascicularis , Masculino , Microsomas/metabolismo , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Ratas , Ratas Gunn , Ratas Sprague-Dawley , Ratas Wistar
11.
Drug Metab Pharmacokinet ; 40: 100412, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34352706

RESUMEN

The predicted contributions of flavin-containing monooxygenase 3 (FMO3) to drug candidate N-oxygenations can be estimated using classic base dissociation constants of the N-containing moiety. In this study, metabolic clearance values in human liver microsomes were experimentally determined for available model drugs. Typical metabolic clearance values (34-96 µL/min/mg protein) at pH 8.4 of trimethylamine, benzydamine, and itopride were two-to fourfold higher than those at pH 7.4. In contrast, the metabolic clearance of control drug midazolam at pH 8.4 was half that at pH 7.4. The ratios of clearance values at pH 8.4 to those at pH 7.4 and the substrate pKa (base) values of reported metabolic N-oxygenation sites of trimethylamine, benzydamine, clomipramine, chlorpromazine, tamoxifen, itopride, loxapine, xanomeline, tozasertib, dasatinib, and clozapine were significantly correlated (r = 0.60, p < 0.05, n = 11). These results suggested that the simple comparison of metabolic clearance values at pH 8.4 and at pH 7.4 could be useful for predicting the contributions of FMO3 to the N-oxygenations of new drug candidates. This method, along with in silico pKa (base) values > 8.4, could prove useful for predicting the contributions of FMO3 to N-oxygenations as part of drug development.


Asunto(s)
Microsomas Hepáticos , Preparaciones Farmacéuticas , Sistema Enzimático del Citocromo P-450 , Humanos , Concentración de Iones de Hidrógeno , Oxigenasas
12.
Drug Metab Dispos ; 38(10): 1806-13, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20584745

RESUMEN

Induction of the cytochrome P450 (P450) enzyme is a major concern in the drug discovery processes. To predict the clinical significance of enzyme induction, it is helpful to investigate pharmacokinetic alterations of a coadministered drug in a suitable animal model. In this study, we focus on the induction of CYP3A, which is involved in the metabolism of approximately 50% of marketed drugs and is inducible in both the liver and intestine. As a marker substrate for CYP3A activity, alprazolam (APZ) was selected and characterized using recombinant CYP3A enzymes expressed in Escherichia coli. Both human CYP3A4 and its cynomolgus P450 ortholog predominantly catalyzed APZ 4-hydroxylation with sigmoidal kinetics. When administered intravenously and orally to cynomolgus monkeys, APZ had moderate clearance; its first-pass extraction ratio after oral dosing was estimated to be 0.09 in the liver and 0.45 in the intestine. Pretreatment with multiple doses of rifampicin (20 mg/kg p.o. for 5 days), a known CYP3A inducer, significantly decreased plasma concentrations of APZ after intravenous and oral administrations (0.5 mg/kg), and first-pass extraction ratios were increased to 0.39 in the liver and 0.63 in the intestine. The results were comparable to those obtained in clinical drug-drug interaction (DDI) reports related to CYP3A induction, although the rate of recovery of CYP3A activity seemed to be slower than rates estimated in clinical studies. In conclusion, pharmacokinetic studies using APZ as a probe in monkeys may provide useful information regarding the prediction of clinical DDIs due to CYP3A induction.


Asunto(s)
Alprazolam/farmacocinética , Citocromo P-450 CYP3A/biosíntesis , Intestino Delgado/enzimología , Hígado/enzimología , Administración Oral , Alprazolam/administración & dosificación , Alprazolam/sangre , Alprazolam/metabolismo , Animales , Citocromo P-450 CYP3A/genética , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Inducción Enzimática , Escherichia coli/enzimología , Escherichia coli/genética , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Inyecciones Intravenosas , Intestino Delgado/metabolismo , Hígado/metabolismo , Macaca fascicularis , Masculino , Tasa de Depuración Metabólica , Microsomas/enzimología , Microsomas/metabolismo , Modelos Animales , Valor Predictivo de las Pruebas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transfección
13.
Drug Metab Dispos ; 37(11): 2127-36, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19704026

RESUMEN

Irreversible inhibition, characterized as mechanism-based inhibition (MBI), of cytochrome P450 in drugs has to be avoided for their safe use. A comprehensive assessment of drug-drug interaction (DDI) potential is important during the drug discovery process. In the present study, we evaluated the effects of macrolide antibiotics, erythromycin (ERM), clarithromycin (CAM), and azithromycin (AZM), which are mechanism-based inhibitors of CYP3A, on biotransformation of midazolam (MDZ) in monkeys. These macrolides inhibited the formation of 1'-hydroxymidazolam in monkey microsomes as functions of incubation time and macrolide concentration. Furthermore, the inactivation potentials of macrolides (k(inact)/K(I): CAM congruent with ERM > AZM) were as effective as that observed in human samples. In in vivo studies, MDZ was administered orally (1 mg/kg) without or with multiple oral dosing of macrolides (15 mg/kg, twice a day on days 1-3). On day 3, the area under the plasma concentration-time curve (AUC) of MDZ increased 7.0-, 9.9-, and 2.0-fold with ERM, CAM, and AZM, respectively, compared with MDZ alone. Furthermore, the effects of ERM and CAM on the pharmacokinetics of MDZ were also observed on the day (day 4) after completion of macrolide treatments (AUC changes: 7.3- and 7.3-fold, respectively). Because the plasma concentrations of macrolides immediately before MDZ administration on day 4 were much lower than the IC(50) values for reversible CYP3A inhibition, the persistent effects may be predominantly caused by CYP3A inactivation. These results suggest that the monkey might be a suitable animal model to predict DDIs caused by MBI of CYP3A.


Asunto(s)
Antibacterianos/metabolismo , Antibacterianos/farmacología , Macrólidos/metabolismo , Macrólidos/farmacología , Animales , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Interacciones Farmacológicas/fisiología , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Macaca fascicularis , Masculino , Microsomas/efectos de los fármacos , Microsomas/enzimología
14.
Drug Metab Dispos ; 37(1): 122-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18854378

RESUMEN

Drugs with potential drug-drug interactions (DDIs) may have a limited scope of use and, at worst, may have to be withdrawn from the market. Therefore, during the drug discovery process it is important to select drug candidates with reduced potential for DDIs. In the present study, we evaluated the pharmacokinetics of simvastatin (SV), a typical substrate for cytochrome P450 (P450) 3A, and examined the DDI between SV and ketoconazole (KTZ), a P450 3A inhibitor, in monkeys. SV metabolism in monkey liver and intestinal microsomes was almost completely inhibited by addition of anti-P450 3A4 antiserum. A similar effect was seen in human microsomes, and the IC(50) values of KTZ for inhibition of SV metabolism were similar in monkey and human samples. In vivo, there were no significant differences in the pharmacokinetic parameters of SV and SVA after i.v. administration of SV in the presence of KTZ compared with those in controls, probably because of the limited systemic exposure to KTZ. In contrast, the pharmacokinetics of SV and SVA after p.o. administration of SV were significantly influenced by the presence of KTZ, and C(max) and area under the plasma concentration-time curve were approximately 5 to 10 times higher than those after p.o. dosing with SV alone. The increases in systemic SV exposure caused by a concomitant p.o. dose of KTZ in monkeys were similar to those observed in clinical studies, which suggests that monkeys might be a suitable animal model in which to predict DDIs involving P450 3A inhibition.


Asunto(s)
Antifúngicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Cetoconazol/farmacología , Simvastatina/farmacocinética , Administración Oral , Animales , Antifúngicos/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Infusiones Intravenosas , Cetoconazol/administración & dosificación , Macaca fascicularis , Masculino , Espectrometría de Masas , Simvastatina/administración & dosificación
15.
Drug Metab Pharmacokinet ; 34(4): 233-238, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31176593

RESUMEN

Creatinine is excreted into urine by glomerular filtration and renal tubular secretion through drug transporters such as organic anion transporter 2 (OAT2), organic cation transporter 2 (OCT2), OCT3, multidrug and toxin extrusion protein 1 (MATE1), and MATE2-K. We aimed to investigate whether our method for estimating percentage changes in serum creatinine concentration (SCr) and creatinine clearance (CLcre) from the baseline is applicable for studying renal transporter inhibitors. We tested 14 compounds (cimetidine, cobicistat, dolutegravir, dronedarone, DX-619, famotidine, INCB039110, nizatidine, ondansetron, pyrimethamine, rabeprazole, ranolazine, trimethoprim, and vandetanib), which were reported to cause reversible changes in SCr and/or CLcre in healthy subjects excluding elderly. Percentage changes were estimated from the relative contributions of the forementioned transporters to CLcre and competitive inhibition by these compounds at their maximum plasma unbound concentrations. For 7 and 9 out of these compounds, changes in SCr and/or CLcre were estimated within 2- and 3-fold of observed values, respectively. Less than 10% changes in SCr and/or CLcre caused by cobicistat, dolutegravir, and rabeprazole were reproduced as such by our method. These findings suggest that our method can be used to estimate changes in SCr and CLcre caused by competitive inhibitions of renal drug transporters.


Asunto(s)
Creatinina/sangre , Creatinina/farmacocinética , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Inhibidores de la Bomba de Protones/farmacología , Transporte Biológico/efectos de los fármacos , Voluntarios Sanos , Humanos , Proteínas de Transporte de Catión Orgánico/metabolismo
16.
Drug Metab Pharmacokinet ; 33(1): 103-110, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29361388

RESUMEN

Serum creatinine (SCr) levels rise during trimethoprim therapy for infectious diseases. This study aimed to investigate whether the elevation of SCr can be quantitatively explained using a physiologically-based pharmacokinetic (PBPK) model incorporating inhibition by trimethoprim on tubular secretion of creatinine via renal transporters such as organic cation transporter 2 (OCT2), OCT3, multidrug and toxin extrusion protein 1 (MATE1), and MATE2-K. Firstly, pharmacokinetic parameters in the PBPK model of trimethoprim were determined to reproduce the blood concentration profile after a single intravenous and oral administration of trimethoprim in healthy subjects. The model was verified with datasets of both cumulative urinary excretions after a single administration and the blood concentration profile after repeated oral administration. The pharmacokinetic model of creatinine consisted of the creatinine synthesis rate, distribution volume, and creatinine clearance (CLcre), including tubular secretion via each transporter. When combining the models for trimethoprim and creatinine, the predicted increments in SCr from baseline were 29.0%, 39.5%, and 25.8% at trimethoprim dosages of 5 mg/kg (b.i.d.), 5 mg/kg (q.i.d.), and 200 mg (b.i.d.), respectively, which were comparable with the observed values. The present model analysis enabled us to quantitatively explain increments in SCr during trimethoprim treatment by its inhibition of renal transporters.


Asunto(s)
Creatinina/sangre , Inhibidores del Citocromo P-450 CYP2C8/sangre , Riñón/metabolismo , Modelos Biológicos , Trimetoprim/sangre , Inhibidores del Citocromo P-450 CYP2C8/farmacología , Voluntarios Sanos , Humanos , Riñón/efectos de los fármacos , Trimetoprim/farmacología
17.
Drug Metab Pharmacokinet ; 32(3): 179-188, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28412023

RESUMEN

The constitutive androstane receptor (CAR, NR1I3) regulates the expression of numerous drug-metabolizing enzymes and transporters. The upregulation of various enzymes, including CYP2B6, by CAR activators is a critical problem leading to clinically severe drug-drug interactions (DDIs). To date, however, few effective computational approaches for identifying CAR activators exist. In this study, we aimed to develop three-dimensional quantitative structure-activity relationship (3D-QSAR) models to predict the CAR activating potency of compounds emerging in the drug-discovery process. Models were constructed using comparative molecular field analysis (CoMFA) based on the molecular alignments of ligands binding to CAR, which were obtained from ensemble ligand-docking using 28 compounds as a training set. The CoMFA model, modified by adding a lipophilic parameter with calculated logD7.4 (S+logD7.4), demonstrated statistically good predictive ability (r2 = 0.99, q2 = 0.74). We also confirmed the excellent predictability of the 3D-QSAR model for CAR activation (r2pred = 0.71) using seven compounds as a test set for external validation. Collectively, our results indicate that the 3D-QSAR model developed in this study provides precise prediction of CAR activating potency and, thus, should be useful for selecting drug candidates with minimized DDI risk related to enzyme-induction in the early drug-discovery stage.


Asunto(s)
Relación Estructura-Actividad Cuantitativa , Receptores Citoplasmáticos y Nucleares/química , Receptor de Androstano Constitutivo , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular
18.
Drug Metab Pharmacokinet ; 20(3): 206-11, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15988123

RESUMEN

The metabolism of ethyl 2-(4-chlorophenyl)-5-(2-furyl)-4-oxazoleacetate (TA-1801), a potent hypolipidemic agent, was studied in humans after oral administration and compared with that found in rats, rabbits, and dogs previously. Hydrolysis of the ethyl ester to produce metabolite M1 (TA-1801 active form; TA-1801A) is the first metabolic step and the subsequent biotransformation includes the glucuronidation to form the metabolite M4 and the oxidation to form the metabolites M2 and M3. The metabolism of TA-1801 in humans was qualitatively similar to that in the experimental animals studied, although species differences were seen in the amount of metabolites. M4, the glucuronide of TA-1801A was the most abundant metabolite in human urine (24.3% of the dose). In vitro studies using human liver and jejunum microsomes indicated that the TA-1801A glucuronosyltransferase activity in human jejunum microsomes was 2-fold higher than that in liver microsomes. With regard to the interspecies differences in the TA-1801A glucuronosyltransferase activities, the intrinsic clearance for the TA-1801A glucuronidation in liver microsomes was in the following order: rabbit>monkey>human=rat=dog. In jejunum microsomes, the intrinsic clearance for the TA-1801A glucuronidation was in the following order: human>monkey>rabbit>rat=dog. These results suggest that the species differences in the intestinal TA-1801A glucuronidation contribute to the species differences in the excretion rate of TA-1801A glucuronide into the urine.


Asunto(s)
Glucuronatos/metabolismo , Yeyuno/metabolismo , Microsomas Hepáticos/metabolismo , Oxazoles/metabolismo , Adulto , Animales , Perros , Glucuronosiltransferasa/metabolismo , Humanos , Yeyuno/citología , Masculino , Persona de Mediana Edad , Oxazoles/orina , Conejos , Ratas , Especificidad de la Especie
19.
Drug Metab Pharmacokinet ; 20(3): 212-8, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15988124

RESUMEN

We characterized the hepatic and intestinal UDP-glucuronosyltransferase (UGT) isoform(s) responsible for the glucuronidation of 2-(4-chlorophenyl)-5-(2-furyl)-4-oxazoleacetic acid (TA-1801A) in humans through several in vitro mechanistic studies. Assessment of a panel of recombinant UGT isoforms revealed the TA-1801A glucuronosyltransferase activity of UGT1A1, UGT1A3, UGT1A7, UGT1A9, and UGT2B7. Kinetic analyses of the TA-1801A glucuronidation by recombinant UGT1A1, UGT1A3, UGT1A9, and UGT2B7 showed that the K(m) value for UGT2B7 was apparently consistent with those in human liver and jejunum microsomes. The TA-1801A glucuronosyltransferase activity in human liver microsomes was inhibited by bilirubin (typical substrate for UGT1A1), propofol (typical substrate for UGT1A9), diclofenac (substrate for UGT1A9 and UGT2B7), and genistein (substrate for UGT1A1, UGT1A3, and UGT1A9). The inhibition by bilirubin, propofol, and diclofenac of the TA-1801A glucuronidation was less pronounced in jejunum microsomes than liver microsomes, suggesting that the contribution of UGT1A1, UGT1A9, and UGT2B7 to the TA-1801A glucuronidation is smaller in the intestine than the liver. In contrast, genistein strongly inhibited the TA-1801A glucuronosyltransferase activity in both human liver and jejunum microsomes. These results suggest that the glucuronidation of TA-1801A is mainly catalyzed by UGT1A1, UGT1A9, and UGT2B7 in the liver, and by UGT1A1, UGT1A3, and UGT2B7 in the intestine in humans.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Isoenzimas/metabolismo , Oxazoles/metabolismo , Glucuronosiltransferasa/antagonistas & inhibidores , Humanos , Yeyuno/citología , Yeyuno/enzimología , Cinética , Microsomas/enzimología , Microsomas Hepáticos/enzimología , Proteínas Recombinantes/metabolismo
20.
Drug Metab Pharmacokinet ; 20(2): 127-34, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15855725

RESUMEN

The metabolism of a novel dual antagonist for alpha4beta1/alpha4beta7 integrin, TR-14035, and the role of polymorphic enzyme responsible for this metabolism were investigated. Human liver microsomes catalyzed the NADPH-dependent metabolism of TR-14035 to a primary metabolite, O-desmethyl TR-14035. This formation was completely blocked by both sulfaphenazole, a selective CYP2C9 inhibitor, and CYP2C9 antibody, whereas potent inhibitors selective for other CYPs exhibited little effects. Of 12 recombinant CYPs examined, O-desmethyl metabolite was principally formed by CYP2C9. CYP1A1, an extrahepatic enzyme, also had this activity (about one-fourth of CYP2C9). Utilizing recombinant CYP2C9*1, K(m) and V(max)/K(m) values of 23.3 microM and 0.284 microL/min/pmol CYP2C9, respectively, were obtained for the O-desmethyl formation, which were quite similar to those in CYP2C9*2 enzyme. In contrast, V(max)/K(m) value in recombinant CYP2C9*3 was approximately one-sixth of CYP2C9*1 and *2. In agreement, kinetics studies using human liver microsomes with CYP2C9*1/*1, *2/*2 and *3/*3 genotypes revealed that the V(max)/K(m) value in *2/*2 microsomes was comparable to that in wild type microsomes, in contrast, that in *3/*3 microsomes was reduced. These results demonstrate CYP2C9 is a primary enzyme mediating the O-desmethylation of TR-14035 in human liver. In homozygotes of CYP2C9*3, the metabolic clearance of TR-14035 should be decreased compared with homozygotes of CYP2C9*1 or 2.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/fisiología , Integrina alfa4beta1/antagonistas & inhibidores , Integrinas/antagonistas & inhibidores , Microsomas Hepáticos/enzimología , Fenilalanina/análogos & derivados , Fenilalanina/farmacocinética , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Hidrocarburo de Aril Hidroxilasas/genética , Citocromo P-450 CYP2C9 , Humanos , Técnicas In Vitro , Isoenzimas/fisiología , Polimorfismo Genético , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA