Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Chem Commun (Camb) ; 59(34): 5071-5074, 2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-37021390

RESUMO

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.


Assuntos
Doenças não Transmissíveis , Humanos , Corantes Fluorescentes/química , RNA/química , Espectrometria de Fluorescência
2.
Drug Metab Pharmacokinet ; 40: 100412, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34352706

RESUMO

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.


Assuntos
Microssomos Hepáticos , Preparações Farmacêuticas , Sistema Enzimático do Citocromo P-450 , Humanos , Concentração de Íons de Hidrogênio , Oxigenases
3.
Drug Metab Pharmacokinet ; 34(4): 233-238, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31176593

RESUMO

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.


Assuntos
Creatinina/sangue , Creatinina/farmacocinética , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Inibidores da Bomba de Prótons/farmacologia , Transporte Biológico/efeitos dos fármacos , Voluntários Saudáveis , Humanos , Proteínas de Transporte de Cátions Orgânicos/metabolismo
4.
Biopharm Drug Dispos ; 40(2): 62-69, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30652318

RESUMO

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.


Assuntos
Transportadores de Ânions Orgânicos/metabolismo , Quinolinas/metabolismo , Proteínas Recombinantes/metabolismo , Telmisartan/metabolismo , Animais , Interações Medicamentosas , Células HEK293 , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado/genética , Transportador 1 de Ânion Orgânico Específico do Fígado/metabolismo , Macaca fascicularis , Macaca mulatta , Transportadores de Ânions Orgânicos/genética , Plasmídeos , Pravastatina/metabolismo , Proteínas Recombinantes/genética , Rosuvastatina Cálcica/metabolismo , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/genética , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/metabolismo , Especificidade por Substrato
5.
Drug Metab Pharmacokinet ; 33(1): 103-110, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29361388

RESUMO

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.


Assuntos
Creatinina/sangue , Inibidores do Citocromo P-450 CYP2C8/sangue , Rim/metabolismo , Modelos Biológicos , Trimetoprima/sangue , Inibidores do Citocromo P-450 CYP2C8/farmacologia , Voluntários Saudáveis , Humanos , Rim/efeitos dos fármacos , Trimetoprima/farmacologia
6.
Drug Metab Dispos ; 45(9): 1027-1034, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28698303

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Glucuronosiltransferase/metabolismo , Intestinos/enzimologia , Adulto , Idoso , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Feminino , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/imunologia , Células HEK293 , Células Hep G2 , Humanos , Intestinos/citologia , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley
7.
Drug Metab Pharmacokinet ; 32(3): 179-188, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28412023

RESUMO

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.


Assuntos
Relação Quantitativa Estrutura-Atividade , Receptores Citoplasmáticos e Nucleares/química , Receptor Constitutivo de Androstano , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular
8.
Biopharm Drug Dispos ; 37(8): 479-490, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27603548

RESUMO

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.


Assuntos
Antifúngicos/metabolismo , Ciclosporina/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/metabolismo , Transportador 1 de Ânion Orgânico Específico do Fígado/biossíntese , Transportadores de Ânions Orgânicos Sódio-Independentes/biossíntese , Quinolinas/metabolismo , Animais , Antifúngicos/farmacologia , Transporte Biológico/fisiologia , Ciclosporina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas/fisiologia , Células HEK293 , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Macaca fascicularis , Quinolinas/farmacologia , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto
9.
Drug Metab Dispos ; 44(7): 888-96, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27112166

RESUMO

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.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Rim/efeitos dos fármacos , Tolmetino/análogos & derivados , Injúria Renal Aguda/sangue , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Animais , Antioxidantes/farmacologia , Biomarcadores/sangue , Biotransformação , Nitrogênio da Ureia Sanguínea , Butionina Sulfoximina/farmacologia , Creatinina/sangue , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Mediadores da Inflamação/metabolismo , Rim/metabolismo , Rim/patologia , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Marcadores de Spin , Fatores de Tempo , Tolmetino/administração & dosagem , Tolmetino/sangue , Tolmetino/toxicidade , Tritolil Fosfatos/farmacologia
10.
Biopharm Drug Dispos ; 37(3): 142-155, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26749565

RESUMO

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.

11.
Toxicol In Vitro ; 30(1 Pt B): 241-9, 2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26528891

RESUMO

Chemical reactivity of acyl glucuronides (AGs) is believed to be involved in the toxicity of carboxylic acid-containing drugs. Both direct and immune-mediated toxicity have been suggested as possible mechanisms of toxicity; however, it remains unclear. In the present study, we performed assays of half-lives, peptide adducts, and immunostimulation to evaluate the potential risk of AGs of 21 drugs and analyzed the relationship to the toxic category. AGs of all withdrawn drugs tested in this study showed short half-lives and peptide adducts formation, but so did those of several safe drugs. In contrast, only AGs of withdrawn and warning drugs induced interleukin-8 (IL-8) in human peripheral blood mononuclear cells (hPBMCs). Using a DNA microarray assay, we found that zomepirac AG induced the mRNAs of 5 genes, including IL-8 in hPBMCs. In addition, withdrawn and warning drugs were distinguished from safe drugs by an integrated score of relative mRNA expression levels of 5 genes. The immunostimulation assay showed higher sensitivity, specificity, and accuracy compared with other methods. In preclinical drug development, the evaluation of the reactivity of AGs using half-lives and peptide adducts assays followed by the evaluation of immunostimulation by highly reactive AGs using hPBMCs can contribute to improved drug safety.


Assuntos
Glucuronídeos/toxicidade , Peptídeos/metabolismo , Células Cultivadas , Glucuronídeos/farmacocinética , Meia-Vida , Humanos , Imunização , Interleucina-8/genética , RNA Mensageiro/análise
12.
Drug Metab Pharmacokinet ; 30(1): 64-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25760531

RESUMO

Benzydamine is an anti-inflammatory drug that undergoes flavin-containing monooxygenase (FMO)-dependent metabolism to benzydamine N-oxide; however, benzydamine N-demethylation is also catalyzed by liver microsomes. In this study, benzydamine N-oxygenation and N-demethylation mediated by liver microsomes from rats, dogs, monkeys, and humans were characterized comprehensively. Values of the maximum velocity/Michaelis constant ratio for benzydamine N-oxygenation by liver microsomes from dogs and rats were higher than those from monkeys and humans, despite roughly similar rates of N-demethylation in the four species. Benzydamine N-oxygenation by liver microsomes was extensively suppressed by preheating liver microsomes at 45 °C for 5 min or at 37 °C for 5-10 min without NADPH, and benzydamine N-demethylation was strongly inhibited by 1-aminbobenztriazole. Liver microsomal benzydamine N-oxygenation was inhibited by dimethyl sulfoxide and methimazole, whereas N-demethylation was inhibited by quinidine. High benzydamine N-oxygenation activities of recombinant human FMO1 and FMO3 and human kidney microsomes were observed at pH 8.4, whereas N-demethylation by cytochrome P450 2D6 was faster at pH 7.4. These results suggest that benzydamine N-oxygenation and N-demethylation are mediated by FMO1/3 and P450s, respectively, and that the contribution of FMO to metabolic eliminations of new drug candidates might be underestimated under certain experimental conditions suitable for P450 enzymes.


Assuntos
Anti-Inflamatórios/metabolismo , Benzidamina/análogos & derivados , Sistema Enzimático do Citocromo P-450/metabolismo , Microssomos Hepáticos/metabolismo , Oxigenases/metabolismo , Animais , Benzidamina/metabolismo , Biotransformação , Sistema Enzimático do Citocromo P-450/genética , Cães , Humanos , Técnicas In Vitro , Macaca fascicularis , Masculino , Metilação , Microssomos Hepáticos/enzimologia , Oxirredução , Oxigenases/genética , Ratos Sprague-Dawley , Proteínas Recombinantes , Especificidade da Espécie
13.
Toxicol Lett ; 232(1): 79-88, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25455449

RESUMO

Phenytoin, 5,5-diphenylhydantoin (DPH), is widely used as an anticonvulsant agent. Severe hepatic injury rarely occurs in patients who received DPH. The development of liver injury is thought to be caused by reactive metabolites; however, the metabolites suggested to contribute to hepatotoxicity have not yet been detected in vivo and their effect on developing the liver injury is largely unknown. We recently demonstrated that DPH treatment decreased hepatic glutathione (GSH) contents, and GSH-depleted condition exacerbated DPH-induced liver injury in mice. The aim of the present study was to identify the reactive metabolite and to investigate the role of P450-mediated metabolisms in DPH-induced liver injury. We identified a novel GSH-conjugated (GS)-DPH, a conjugate of putative electrophilic arene oxide intermediate with GSH, in the bile of mice with DPH-induced liver injury. In plasma, cysteine- or N-acetylcysteine-conjugated DPH was detected, and these thiol conjugates levels were correlated with the plasma alanine aminotransferase (ALT) levels. These changes were significantly reduced by pretreatment with P450 inhibitor. Furthermore, the increases of hepatic P450 activities were in parallel with elevation of plasma thiol conjugates levels. These findings suggest that the arene oxide intermediate, which can be converted to thiol conjugates, is involved in DPH-induced liver injury.


Assuntos
Anticonvulsivantes/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/enzimologia , Fenitoína/metabolismo , Compostos de Sulfidrila/metabolismo , Acetilcisteína/metabolismo , Ativação Metabólica , Alanina Transaminase/sangue , Animais , Bile/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450/farmacologia , Modelos Animais de Doenças , Feminino , Glutationa/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos Endogâmicos C57BL , Compostos de Sulfidrila/sangue , Fatores de Tempo
14.
Xenobiotica ; 45(6): 556-62, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25539456

RESUMO

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.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Glucuronatos/farmacocinética , Fígado/metabolismo , Piperidinas/farmacocinética , Valsartana/farmacocinética , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Estradiol/análogos & derivados , Estradiol/farmacocinética , Estradiol/farmacologia , Glucuronatos/farmacologia , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/metabolismo , Masculino , Piperidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Valsartana/farmacologia
15.
Xenobiotica ; 44(3): 242-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23855261

RESUMO

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.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Inibidores da Dipeptidil Peptidase IV/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacocinética , Oxigenases/metabolismo , Pirazóis/metabolismo , Pirazóis/farmacocinética , Tiazolidinas/metabolismo , Tiazolidinas/farmacocinética , Absorção , Adulto , Área Sob a Curva , Radioisótopos de Carbono/sangue , Radioisótopos de Carbono/farmacocinética , Cromatografia Líquida de Alta Pressão , Inibidores da Dipeptidil Peptidase IV/sangue , Inibidores da Dipeptidil Peptidase IV/química , Fezes/química , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Modelos Biológicos , Estrutura Molecular , Pirazóis/sangue , Pirazóis/química , Contagem de Cintilação , Tiazolidinas/sangue , Tiazolidinas/química , Fatores de Tempo
16.
Drug Metab Dispos ; 41(10): 1875-82, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23929936

RESUMO

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.


Assuntos
Interações Medicamentosas/genética , Fígado/metabolismo , Macaca fascicularis/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Quinolinas/farmacologia , Quinolinas/farmacocinética , Animais , Área Sob a Curva , Transporte Biológico/genética , Trato Gastrointestinal/metabolismo , Meia-Vida , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Fígado/efeitos dos fármacos , Macaca fascicularis/genética , Masculino , RNA Mensageiro/genética
18.
Drug Metab Dispos ; 39(9): 1495-502, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21646435

RESUMO

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.


Assuntos
Glucuronídeos/metabolismo , Mucosa Intestinal/metabolismo , Cloridrato de Raloxifeno/farmacocinética , Animais , Disponibilidade Biológica , Cães , Descoberta de Drogas/métodos , Glucuronosiltransferase/metabolismo , Fígado/metabolismo , Macaca fascicularis , Masculino , Microssomos/metabolismo , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Ratos , Ratos Gunn , Ratos Sprague-Dawley , Ratos Wistar
19.
Drug Metab Dispos ; 38(10): 1806-13, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20584745

RESUMO

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.


Assuntos
Alprazolam/farmacocinética , Citocromo P-450 CYP3A/biossíntese , Intestino Delgado/enzimologia , Fígado/enzimologia , Administração Oral , Alprazolam/administração & dosagem , Alprazolam/sangue , Alprazolam/metabolismo , Animais , Citocromo P-450 CYP3A/genética , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Indução Enzimática , Escherichia coli/enzimologia , Escherichia coli/genética , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Humanos , Injeções Intravenosas , Intestino Delgado/metabolismo , Fígado/metabolismo , Macaca fascicularis , Masculino , Taxa de Depuração Metabólica , Microssomos/enzimologia , Microssomos/metabolismo , Modelos Animais , Valor Preditivo dos Testes , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transfecção
20.
Drug Metab Dispos ; 37(11): 2127-36, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19704026

RESUMO

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.


Assuntos
Antibacterianos/metabolismo , Antibacterianos/farmacologia , Macrolídeos/metabolismo , Macrolídeos/farmacologia , Animais , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Interações Medicamentosas/fisiologia , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Macaca fascicularis , Masculino , Microssomos/efeitos dos fármacos , Microssomos/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA