Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Drug Metab Dispos ; 48(9): 769-777, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32616542

RESUMEN

Esaxerenone (CS-3150) is a novel, oral, nonsteroidal, selective mineralocorticoid receptor blocker approved for the treatment of hypertension in Japan. Here, the drug-drug interaction (DDI) potential of esaxerenone was evaluated in vitro, and its impact in clinical practice was estimated. Esaxerenone exhibited time-dependent inhibition and induction of CYP3A. When the clinical impacts of esaxerenone on the inhibition and induction of CYP3A were estimated separately by using a mechanistic static model, the predicted area under the curve ratios (AUCRs) of midazolam, a typical CYP3A substrate, were 1.80 and 0.31, respectively, suggesting that the DDI potential of esaxerenone cannot be neglected. Because it was suggested that DDIs mainly occur in the intestine, predictions using concentration-time profiles in each segment of the gastrointestinal tract were performed with GastroPlus, a physiologically based pharmacokinetic (PBPK) modeling software. The predicted AUCR of midazolam was approximately 1.2, which is close to that in a clinical study, despite the difficulty of predicting DDIs for compounds with both inhibition and induction effects. When only inhibition or induction was incorporated into a model, the AUCR of midazolam changed depending on the dosing period and dose level of esaxerenone and the timing of midazolam administration. However, the AUCR calculated by incorporating both effects remained almost constant. This study shows the ability of PBPK models to simulate weak DDIs via intestinal CYP3A and that esaxerenone has low DDI potential as a perpetrator because of the offset of inhibition and induction. SIGNIFICANCE STATEMENT: Weak CYP3A inhibition and/or induction sometimes cause DDIs in the intestine but not the liver. Because strong inhibitors maximally inhibit intestinal CYP3A, the predictability of weak DDIs in the intestine should be evaluated further. Here, we simulate the DDIs of esaxerenone as a perpetrator by using physiologically based pharmacokinetic modeling focusing on the intestine and offset of inhibition and induction.


Asunto(s)
Inductores del Citocromo P-450 CYP3A/farmacocinética , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Antagonistas de Receptores de Mineralocorticoides/farmacocinética , Modelos Biológicos , Pirroles/farmacocinética , Sulfonas/farmacocinética , Administración Oral , Área Bajo la Curva , Simulación por Computador , Citocromo P-450 CYP3A/metabolismo , Inductores del Citocromo P-450 CYP3A/administración & dosificación , Inhibidores del Citocromo P-450 CYP3A/administración & dosificación , Interacciones Farmacológicas , Glucuronosiltransferasa/metabolismo , Humanos , Hipertensión/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/enzimología , Japón , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Microsomas Hepáticos , Midazolam/farmacocinética , Antagonistas de Receptores de Mineralocorticoides/administración & dosificación , Pirroles/administración & dosificación , Medición de Riesgo/métodos , Sulfonas/administración & dosificación
2.
Xenobiotica ; 49(8): 961-969, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30124356

RESUMEN

A 1,2,4-oxadiazole ring-containing compound DS-8500a was developed as a novel G protein-coupled receptor 119 agonist. In vivo metabolic fates of [14C]DS-8500a differently radiolabeled in the benzene ring or benzamide side carbon in rats were investigated. Differences in mass balances were observed, primarily because after the oxadiazole ring-opening and subsequent ring-cleavage small-molecule metabolites containing the benzene side were excreted in the urine, while those containing the benzamide side were excreted in the bile. DS-8500a was detected at trace levels in urine and bile, demonstrating extensive metabolism prior to urinary/biliary excretion. At least 16 metabolite structures were proposed in plasma, urine, and bile samples from rats treated with [14C]DS-8500a. Formation of a ring-opened metabolite (reduced DS-8500a) in hepatocytes of humans, monkeys, and rats was confirmed; however, it was not affected by typical inhibitors of cytochrome P450s, aldehyde oxidases, or carboxylesterases in human hepatocytes. Extensive formation of the ring-opened metabolite was observed in human liver microsomes fortified with an NADPH-generating system under anaerobic conditions. These results suggest an in vivo unique reductive metabolism of DS-8500a is mediated by human non-cytochrome P450 enzymes.


Asunto(s)
Benzamidas/metabolismo , Ciclopropanos/metabolismo , Redes y Vías Metabólicas , Oxadiazoles/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Administración Oral , Anaerobiosis , Animales , Benzamidas/administración & dosificación , Benzamidas/sangre , Benzamidas/farmacocinética , Radioisótopos de Carbono/química , Ciclopropanos/administración & dosificación , Ciclopropanos/sangre , Ciclopropanos/farmacocinética , Humanos , Macaca fascicularis , Masculino , Oxadiazoles/administración & dosificación , Oxadiazoles/sangre , Oxadiazoles/farmacocinética , Oxidación-Reducción , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
3.
Drug Metab Dispos ; 42(4): 520-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24459178

RESUMEN

Edoxaban (the free base of DU-176b), an oral direct factor Xa inhibitor, is mainly excreted unchanged into urine and feces. Because active membrane transport processes such as active renal secretion, biliary excretion, and/or intestinal secretion, and the incomplete absorption of edoxaban after oral administration have been observed, the involvement of drug transporters in the disposition of edoxaban was investigated. Using a bidirectional transport assay in human colon adenocarcinoma Caco-2 cell monolayers, we observed the vectorial transport of [(14)C]edoxaban, which was completely inhibited by verapamil, a strong P-glycoprotein (P-gp) inhibitor. In an in vivo study, an increased distribution of edoxaban to the brain was observed in Mdr1a/1b knockout mice when compared with wild-type mice, indicating that edoxaban is a substrate for P-gp. However, there have been no observations of significant transport of edoxaban by renal or hepatic uptake transporters, organic anion transporter (OAT)1, OAT3, organic cation transporter (OCT)2, or organic anion transporting polypeptide (OATP)1B1. Edoxaban exhibited no remarkable inhibition of OAT1, OAT3, OCT1, OCT2, OATP1B1, OATP1B3, or P-gp up to 30 µM; therefore, the risk of clinical drug-drug interactions due to any edoxaban-related transporter inhibition seems to be negligible. Our results demonstrate that edoxaban is a substrate of P-gp but not of other major uptake transporters tested. Because metabolism is a minor contributor to the total clearance of edoxaban and strong P-gp inhibitors clearly impact edoxaban transport, the P-gp transport system is a key factor for edoxaban's disposition.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Inhibidores del Factor Xa , Piridinas/farmacocinética , Tiazoles/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Transporte Biológico , Células CACO-2 , Células HEK293 , Hepatocitos/metabolismo , Humanos , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Oocitos/metabolismo , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Piridinas/administración & dosificación , Especificidad por Sustrato , Tiazoles/administración & dosificación , Distribución Tisular , Xenopus laevis
4.
Drug Metab Dispos ; 41(5): 1156-62, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23471504

RESUMEN

Olmesartan medoxomil (OM) is a prodrug-type angiotensin II type 1 receptor antagonist. OM is rapidly converted into its active metabolite olmesartan by multiple hydrolases in humans, and we recently identified carboxymethylenebutenolidase homolog (CMBL) as one of the OM bioactivating hydrolases. In the present study, we further investigated the interindividual variability of mRNA and protein expression of CMBL and OM-hydrolase activity using 40 individual human liver and 30 intestinal specimens. In the intestinal samples, OM-hydrolase activity strongly correlated with the CMBL protein expression, clearly indicating that CMBL is a major contributor to the prodrug bioactivation in human intestine. The protein and activity were highly distributed in the proximal region (duodenum and jejunum) and decreased to the distal region of the intestine. Although there was high interindividual variability (16-fold) in both the protein and activity in the intestinal segments from the duodenum to colon, the interindividual variability in the duodenum and jejunum was relatively small (3.0- and 2.4-fold, respectively). In the liver samples, the interindividual variability in the protein and activity was 4.1- and 6.8-fold, respectively. No sex differences in the protein and activity were shown in the human liver or intestine. A genetically engineered Y155C mutant of CMBL, which was caused by a single nucleotide polymorphism rs35489000, showed significantly lower OM-hydrolase activity than the wild-type protein although no minor allele was genotyped in the 40 individual liver specimens.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Intestinos/enzimología , Hígado/enzimología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Hidrolasas de Éster Carboxílico/genética , Cartilla de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , Adulto Joven
5.
Mol Cell Endocrinol ; 299(2): 178-87, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19133314

RESUMEN

Vitamin D receptor (VDR) regulates the expression of many genes involved in mineral metabolism, cellular proliferation, differentiation and drug biotransformation. We studied the expression and activity of VDR and its heterodimerization partner retinoid X receptor-alpha (RXRalpha) in choriocarcinoma trophoblast cell lines BeWo and JEG-3, in comparison with human isolated placental cytotrophoblasts and human full term placenta. We found that VDR and RXRalpha are localised in the human term placenta trophoblast and expressed in isolated cytotrophoblasts. However, we found low expression and no transcriptional activity of VDR in used choriocarcinoma cell lines. The inhibitor of DNA methylation, 5-deoxy-3'-azacytidine, and histone deacetylase inhibitor sodium butyrate partially restored the expression of VDR, suggesting an epigenetic suppression of the gene in choriocarcinoma cells. Differentiation of BeWo cells resulted in up-regulation of VDR mRNA. Finally, we observed a non-genomic effect of 1,25(OH)(2)D(3) in the activation of the extracellular signal-regulated kinase (ERK) signalling pathway in JEG-3 cells. In conclusion, our results suggest an epigenetic repression of VDR gene expression and activity in choriocarcinoma cell lines, and a non-genomic effect of 1,25(OH)(2)D(3) in JEG-3 cells.


Asunto(s)
Coriocarcinoma/metabolismo , Placenta/metabolismo , Receptores de Calcitriol/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Calcitriol/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Separación Celular , Coriocarcinoma/genética , Epigénesis Genética/efectos de los fármacos , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Genoma Humano/genética , Humanos , Luciferasas/metabolismo , Placenta/citología , Placenta/efectos de los fármacos , Embarazo , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Calcitriol/genética , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Activación Transcripcional/efectos de los fármacos , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo
6.
Eur J Pharm Sci ; 46(5): 367-73, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22418036

RESUMEN

The purpose of this study was to validate human small intestinal and colonic tissue mounted in the Ussing chamber as a tool for predicting the oral drug absorption in humans with the main focus on moderately and poorly permeable compounds. The obtained apparent permeability coefficient (P(app)) of eleven test compounds was compared to their fraction absorbed (Fa) in humans taken from the literature. Beside the conventional P(app) a new parameter, the apparent permeability coefficient total (P(app,total)), involving both the apical-to-basolateral permeability and the time-dependent compound accumulation in the tissue was established. The permeability of lucifer yellow (LY), a fluorescent marker of the paracellular pathway and the test compounds showed no obvious differences between small intestine and colon. Furthermore, small intestinal and colonic tissue from a single donor showed similar permeability of both LY and a transcellularly transported compound metoprolol. All test compounds including low molecular weight hydrophilic compounds such as metformin, atenolol, sulpiride and famotidine showed adequate permeability reflecting human Fa values (R(2)=0.87). The P(app) values of digoxin, a P-glycoprotein (P-gp) substrate, were not significantly affected by the addition of verapamil, a P-gp inhibitor. In contrast, the P(app,total) values of digoxin increased approximately threefold in the presence of verapamil. In conclusion, both small intestinal and colonic tissue mounted in the Ussing chamber provide a good opportunity to predict the oral drug absorption rate in humans even for moderately and poorly absorbed compounds. The novel calculation of P(app,total) allows the study of the carrier-mediated drug-drug interactions in human intestine.


Asunto(s)
Colon/metabolismo , Cámaras de Difusión de Cultivos , Absorción Intestinal , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Preparaciones Farmacéuticas/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Administración Oral , Colon/efectos de los fármacos , Interacciones Farmacológicas , Colorantes Fluorescentes/metabolismo , Humanos , Técnicas In Vitro , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Isoquinolinas/metabolismo , Cinética , Permeabilidad , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/química , Reproducibilidad de los Resultados , Supervivencia Tisular
7.
Eur J Pharm Biopharm ; 76(2): 260-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20600890

RESUMEN

The efflux transporters ABCB1 (p-glycoprotein) and ABCC2 (MRP2) play an essential role in the limitation of oral bioavailability of drugs. In the last years, pharmaceutical surfactants like cremophor® EL or polysorbate 80 have been shown to interact with ABCB1. However, the knowledge about their influence on ABCC2 is still limited. In this study, the interactions of the nonionic surfactants cremophor® EL, cremophor® RH 40, polysorbate 80, vitamin E TPGS 1000, pluronic® PE 10300 and sucrose ester L-1695 with both efflux transporters were investigated on cellular level. Cell accumulation studies and transport studies were performed using transfected MDCK II cell models. The influence of ABCC2 inhibiting surfactants on the expression level of ABCC2 was also studied. The investigations showed that cremophor® EL, vitamin E TPGS 1000 and higher concentrations of polysorbate 80 inhibit both transporters. Pluronic® PE 10300 and sucrose ester L-1695 inhibit ABCB1 but not ABCC2. Cremophor® RH 40 only shows inhibitory activity on ABCC2. During the investigated incubation period none of the inhibiting surfactants caused an alteration in ABCC2mRNA or protein expression. These findings indicate that the observed interactions are caused by specific inhibition of the transport activity of ABCC2.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Tensoactivos/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP , Animales , Transporte Biológico , Línea Celular , Perros , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Tensoactivos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA