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.
Artigo em Inglês | MEDLINE | ID: mdl-32616542

RESUMO

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.


Assuntos
Indutores do Citocromo P-450 CYP3A/farmacocinética , Inibidores do Citocromo P-450 CYP3A/farmacocinética , Antagonistas de Receptores de Mineralocorticoides/farmacocinética , Modelos Biológicos , Pirróis/farmacocinética , Sulfonas/farmacocinética , Administração Oral , Área Sob a Curva , Simulação por Computador , Citocromo P-450 CYP3A/metabolismo , Indutores do Citocromo P-450 CYP3A/administração & dosagem , Inibidores do Citocromo P-450 CYP3A/administração & dosagem , Interações Medicamentosas , Glucuronosiltransferase/metabolismo , Humanos , Hipertensão/tratamento farmacológico , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/enzimologia , Japão , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Microssomos Hepáticos , Midazolam/farmacocinética , Antagonistas de Receptores de Mineralocorticoides/administração & dosagem , Pirróis/administração & dosagem , Medição de Risco/métodos , Sulfonas/administração & dosagem
2.
Xenobiotica ; 49(8): 961-969, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30124356

RESUMO

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.


Assuntos
Benzamidas/metabolismo , Ciclopropanos/metabolismo , Redes e Vias Metabólicas , Oxidiazóis/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Administração Oral , Anaerobiose , Animais , Benzamidas/administração & dosagem , Benzamidas/sangue , Benzamidas/farmacocinética , Radioisótopos de Carbono/química , Ciclopropanos/administração & dosagem , Ciclopropanos/sangue , Ciclopropanos/farmacocinética , Humanos , Macaca fascicularis , Masculino , Oxidiazóis/administração & dosagem , Oxidiazóis/sangue , Oxidiazóis/farmacocinética , Oxirredução , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
3.
Drug Metab Dispos ; 42(4): 520-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24459178

RESUMO

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.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Inibidores do Fator Xa , Piridinas/farmacocinética , Tiazóis/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Transporte Biológico , Células CACO-2 , Células HEK293 , Hepatócitos/metabolismo , Humanos , Rim/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Oócitos/metabolismo , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Piridinas/administração & dosagem , Especificidade por Substrato , Tiazóis/administração & dosagem , Distribuição Tecidual , Xenopus laevis
4.
Drug Metab Dispos ; 41(5): 1156-62, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23471504

RESUMO

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.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Intestinos/enzimologia , Fígado/enzimologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Hidrolases de Éster Carboxílico/genética , Primers do DNA , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , Adulto Jovem
5.
Mol Cell Endocrinol ; 299(2): 178-87, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19133314

RESUMO

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.


Assuntos
Coriocarcinoma/metabolismo , Placenta/metabolismo , Receptores de Calcitriol/metabolismo , Caderinas/genética , Caderinas/metabolismo , Calcitriol/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Separação Celular , Coriocarcinoma/genética , Epigênese Genética/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Genoma Humano/genética , Humanos , Luciferases/metabolismo , Placenta/citologia , Placenta/efeitos dos fármacos , Gravidez , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Calcitriol/genética , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo , Ativação Transcricional/efeitos dos fármacos , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
6.
Eur J Pharm Sci ; 46(5): 367-73, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22418036

RESUMO

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.


Assuntos
Colo/metabolismo , Cultura em Câmaras de Difusão , Absorção Intestinal , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Preparações Farmacêuticas/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Administração Oral , Colo/efeitos dos fármacos , Interações Medicamentosas , Corantes Fluorescentes/metabolismo , Humanos , Técnicas In Vitro , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Isoquinolinas/metabolismo , Cinética , Permeabilidade , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/química , Reprodutibilidade dos Testes , Sobrevivência de Tecidos
7.
Eur J Pharm Biopharm ; 76(2): 260-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20600890

RESUMO

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.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Tensoativos/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP , Animais , Transporte Biológico , Linhagem Celular , Cães , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Tensoativos/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa