Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Drug Metab Dispos ; 49(10): 938-946, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34330717

RESUMO

The successful prospective incorporation of in vitro transporter kinetics in physiologically based pharmacokinetic (PBPK) models to describe drug disposition remains challenging. Although determination of scaling factors to extrapolate in vitro to in vivo transporter kinetics has been facilitated by quantitative proteomics, no robust assessment comparing membrane recoveries between different cells/tissues has been made. HEK293 cells overexpressing OCT2, MATE1, and MATE2K or human kidney cortex were homogenized and centrifuged to obtain the total membrane fractions, which were subsequently subjected to liquid-liquid extraction followed by centrifugation and precipitation to isolate plasma membrane fractions. Plasma membrane recoveries determined by quantitation of the marker Na+/K+-ATPase in lysate and plasma membrane fractions were ≤20% but within 3-fold across different cells and tissues. A separate study demonstrated that recoveries are comparable between basolateral and apical membranes of renal proximal tubules, as measured by Na+/K+-ATPase and γ-glutamyl transpeptidase 1, respectively. The plasma membrane expression of OCT2, MATE1, and MATE2K was quantified and relative expression factors (REFs) were determined as the ratio between the tissue and cell concentrations. Corrections using plasma membrane recovery had minimal impact on REF values (<2-fold). In vitro transporter kinetics of metformin were extrapolated to in vivo using the corresponding REFs in a PBPK model. The simulated metformin exposures were within 2-fold of clinical exposure. These results demonstrate that transporter REFs based on plasma membrane expression enable a prediction of transporter-mediated drug disposition. Such REFs may be estimated without the correction of plasma membrane recovery when the same procedure is applied between different matrices. SIGNIFICANCE STATEMENT: Transporter REFs based on plasma membrane expression enable in vitro-in vivo extrapolation of transporter kinetics. Plasma membrane recoveries as determined by the quantification of sodium-potassium adenosine triphosphatase were comparable between the in vitro and in vivo systems used in the present study, and therefore had minimal impact on the transporter REF values.


Assuntos
Metformina/farmacocinética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Transportador 2 de Cátion Orgânico/metabolismo , Transporte Biológico Ativo/fisiologia , Biotransformação/fisiologia , Membrana Celular/metabolismo , Perfilação da Expressão Gênica/métodos , Células HEK293 , Humanos , Hipoglicemiantes/farmacocinética , Taxa de Depuração Metabólica , Modelos Biológicos , Valor Preditivo dos Testes , Proteômica/métodos , Transcriptoma
2.
Xenobiotica ; 47(9): 825-832, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27609543

RESUMO

1. Accurate predictions of clinical transporter-mediated drug-drug interactions (DDI) from in vitro data can be challenging when compounds have poor solubility and/or high nonspecific binding. Additionally, current DDI predictions for compounds with high plasma-protein binding assume that the unbound fraction in plasma is 0.01, if the experimental value is less than 0.01 or cannot be determined. This approach may result in an overestimation of DDI risk. To overcome these challenges, it may be beneficial to conduct inhibition studies under physiologically relevant conditions. 2. Here, IC50 values, determined in the presence of 4% bovine serum albumin approximating human plasma albumin concentrations, were successfully used to predict DDI for uptake transporters, OATP1B1/1B3, OCT1/2, OAT1/3 and MATE1/2K. 3. The IC50 values of reference inhibitors with 4% bovine serum albumin, considered total IC50, were comparable to the predicted values based on nominal IC50 values determined under protein-free conditions and unbound fraction in plasma. Calculation of R-total and Cmax/IC50,total values using total plasma exposure and total IC50 values explained the clinical DDI or absence of it for these inhibitors. 4. These results suggest that IC50 determinations in the presence of 4% albumin can be used, in the context of clinical total exposure, to predict DDI involving uptake transporters.


Assuntos
Interações Medicamentosas , Concentração Inibidora 50 , Proteínas de Membrana Transportadoras/metabolismo , Humanos , Transportadores de Ânions Orgânicos/metabolismo
3.
Xenobiotica ; 44(3): 276-82, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23886114

RESUMO

1. Transient benign unconjugated hyperbilirubinemia has been observed clinically with several drugs including indinavir, cyclosporine, and rifamycin SV. Genome-wide association studies have shown significant association of OATP1B1 and UGT1A1 with elevations of unconjugated bilirubin, and OATP1B1 inhibition data correlated with clinical unconjugated hyperbilirubinemia for several compounds. 2. In this study, inhibition of OATP1B3 and UGT1A1, in addition to OATP1B1, was explored to determine whether one measure offers value over the other as a potential prospective tool to predict unconjugated hyperbilirubinemia. OATP1B1 and OATP1B3-mediated transport of bilirubin was confirmed and inhibition was determined for atazanavir, rifampicin, indinavir, amprenavir, cyclosporine, rifamycin SV and saquinavir. To investigate the intrinsic inhibition by the drugs, both in vivo Fi (fraction of intrinsic inhibition) and R-value (estimated maximum in vivo inhibition) for OATP1B1, OATP1B3 and UGT1A1 were calculated. 3. The results indicated that in vivo Fi values >0.2 or R-values >1.5 for OATP1B1 or OATP1B3, but not UGT1A1, are associated with previously reported clinical cases of drug-induced unconjugated hyperbilirubinemia. 4. In conclusion, inhibition of OATP1B1 and/or OATP1B3 along with predicted human pharmacokinetic data could be used pre-clinically to predict potential drug-induced benign unconjugated hyperbilirubinemia in the clinic.


Assuntos
Antirreumáticos/farmacologia , Inibidores da Protease de HIV/farmacologia , Hiperbilirrubinemia/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/antagonistas & inibidores , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Sulfato de Atazanavir , Bilirrubina/metabolismo , Carbamatos , Ciclosporina , Furanos , Glucuronosiltransferase/antagonistas & inibidores , Técnicas In Vitro , Indinavir , Transportador 1 de Ânion Orgânico Específico do Fígado , Oligopeptídeos , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Piridinas , Rifampina , Rifamicinas , Saquinavir , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto , Sulfonamidas
4.
Artigo em Inglês | MEDLINE | ID: mdl-19119084

RESUMO

Abnormal elevation of 11beta-HSD1 activities in tissues, such as fat and brain, may contribute to the development of the abdominal obesity and Alzheimer disease, and the inhibition of 11beta-HSD1 might be beneficial to the management of these diseases. To assess the effects of pharmacologic inhibitors of 11beta-HSD1, we developed a fast LC/MS/MS method to quantify corticosteroids in minced tissue samples in the presence of 11beta-HSD substrates. The novel on-line SPE-LC/MS/MS method was developed with dual binary gradient and a throughput of 4.5 min/sample. A total of six corticosteroids (cortisol, cortisone, corticosterone, dehydrocorticosterone, dexamethasone, and dehydrodexamethasone) were studied. The lower limit of quantitation from 0.40 to 11.4 fmol and 4.5 orders magnitude of dynamic range were obtained for these six compounds. Three novel enzymatic bi-products, all isomers of cortisol, were observed in the liver or fat samples. Two of them were identified by matching the HPLC retention times and MS/MS spectra with authentic compounds. The potential interferences of these isomers and their removal are discussed.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/metabolismo , Corticosteroides/análise , Cromatografia Líquida , Extração em Fase Sólida , Espectrometria de Massas em Tandem , 11-beta-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Corticosteroides/metabolismo , Animais , Química Encefálica , Epididimo/química , Desenho de Equipamento , Rim/química , Modelos Lineares , Fígado/química , Masculino , Camundongos , Sensibilidade e Especificidade
5.
J Pharm Sci ; 108(9): 3118-3123, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31034908

RESUMO

Cobicistat has been reported to increase serum creatinine clinically without affecting glomerular filtration. This was ascribed to transient inhibition of MATE1-mediated renal creatinine secretion. Interestingly, a structurally similar drug, ritonavir, has not been associated with serum creatinine increases at the pharmacoenhancer dose. The present study was aimed to investigate the translation of in vitro MATE1/2K inhibition to clinical creatinine increase (cobicistat) and lack of it (ritonavir) considering their intracellular concentrations in renal proximal tubules. Uptake studies showed ritonavir and cobicistat are unlikely substrates for OCT2. The steady-state unbound concentration in the cytosol of human renal proximal tubule epithelial cells was comparable with the extracellular unbound concentration, suggesting that the entry of these compounds is predominantly mediated by passive diffusion. Ritonavir and cobicistat are MATE1 and MATE2K inhibitors with IC50 values of 3.1 and 90 µM (ritonavir), and 4.4 and 3.2 µM (cobicistat), respectively. However, the unbound cytosolic concentrations (Cu,cytosol) of ritonavir and cobicistat in human renal proximal tubule epithelial cells, 0.065 and 0.10 µM, respectively, after incubation with the clinical maximum total plasma concentrations at pharmacoenhancer doses does not support inhibition in vivo; Cu,cytosol >30 fold lower than IC50s. These results demonstrate that MATE1/2K inhibition is unlikely the mechanism of the clinical creatinine elevations with cobicistat.


Assuntos
Cobicistat/farmacologia , Creatinina/metabolismo , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Ritonavir/farmacologia , Linhagem Celular , Creatinina/sangue , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Taxa de Filtração Glomerular/efeitos dos fármacos , Células HEK293 , Humanos , Concentração Inibidora 50 , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Eliminação Renal/efeitos dos fármacos
6.
J Med Chem ; 50(1): 149-64, 2007 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-17201418

RESUMO

Starting from a rapidly metabolized adamantane 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) inhibitor 22a, a series of E-5-hydroxy-2-adamantamine inhibitors, exemplified by 22d and (+/-)-22f, was discovered. Many of these compounds are potent inhibitors of 11beta-HSD1 and are selective over 11beta-HSD2 for multiple species (human, mouse, and rat), unlike other reported species-selective series. These compounds have good cellular potency and improved microsomal stability. Pharmacokinetic profiling in rodents indicated moderate to large volumes of distribution, short half-lives, and a pharmacokinetic species difference with the greatest exposure measured in rat with 22d. One hour postdose liver, adipose, and brain tissue 11beta-HSD1 inhibition was confirmed with (+/-)-22f in a murine ex vivo assay. Although 5,7-disubstitued-2-adamantamines provided greater stability, a single, E-5-position, polar functional group afforded inhibitors with the best combination of stability, potency, and selectivity. These results indicate that adamantane metabolic stabilization sufficient to obtain short-acting, potent, and selective 11beta-HSD1 inhibitors has been discovered.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , Adamantano/análogos & derivados , Adamantano/síntese química , Piperazinas/síntese química , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Adamantano/farmacocinética , Animais , Linhagem Celular , Humanos , Técnicas In Vitro , Camundongos , Microssomos Hepáticos/metabolismo , Piperazinas/farmacocinética , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Distribuição Tecidual
7.
J Pharm Sci ; 102(12): 4426-32, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24122511

RESUMO

Veliparib (ABT-888) is largely eliminated as parent drug in human urine (70% of the dose). Renal unbound clearance exceeds glomerular filtration rate, suggesting the involvement of transporter-mediated active secretion. Clinically relevant pharmacokinetic interactions in the kidney have been associated with OAT1, OAT3, OCT2, MATE1, and MATE2K. In the present study, interactions of veliparib with these transporters were investigated. Veliparib inhibited OAT1, OAT3, OCT2, MATE1, and MATE2K with IC50 values of 1371, 505, 3913, 69.9, and 69.5 µM, respectively. The clinical unbound maximum plasma concentration of veliparib after single oral dose of 50 mg (0.45 µM) is manyfold lower than IC50 values for OAT1, OAT3, OCT2, MATE1, or MATE2K. These results indicate a low potential for drug-drug interaction (DDI) with OAT1/3, OCT2, or MATE1/2K. Additional studies demonstrated that veliparib is a substrate of OCT2. In Oct1/Oct2 double-knockout mice, the plasma exposure of veliparib was increased by 1.5-fold, and the renal clearance was decreased by 1.8-fold as compared with wild-type mice, demonstrating that organic cation transporters contribute to the renal elimination in vivo. In summary, the in vitro transporter data for veliparib predicts minimal potential for an OAT1/3-, OCT2-, and MATE1/2K-mediated DDI given the clinical exposure after single oral dose of 50 mg.


Assuntos
Benzimidazóis/metabolismo , Benzimidazóis/farmacocinética , Rim/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Animais , Benzimidazóis/sangue , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Transportadores de Ânions Orgânicos Sódio-Independentes/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/genética
8.
Clin Sci (Lond) ; 103 Suppl 48: 107S-111S, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12193066

RESUMO

Endothelins (ETs), 21-amino-acid peptides involved in the pathogenesis of various diseases, bind to ET(A) and ET(B) receptors to initiate their effects. Based on the same core structure, we have developed four small-molecule ET receptor antagonists, ABT-627, ABT-546, A-182086 and A-192621, which exhibit difference in selectivity for ET(A) and ET(B) receptors. In this report, we compare the potency and selectivity of these four antagonists in inhibiting (125)I-labelled ET-1 binding to cloned human ET(A) and ET(B) receptors, and in blocking ET-1-induced functional responses (arachidonic acid release and phosphatidylinositol hydrolysis).


Assuntos
Antagonistas dos Receptores de Endotelina , Vasodilatadores/farmacologia , Animais , Ácido Araquidônico/metabolismo , Atrasentana , Ligação Competitiva , Células CHO , Linhagem Celular , Cricetinae , Endotelina-1/metabolismo , Endotelina-1/farmacologia , Humanos , Hidrólise , Radioisótopos do Iodo/metabolismo , Fosfatidilinositóis/metabolismo , Pirrolidinas/farmacologia , Receptor de Endotelina A , Receptor de Endotelina B , Receptores de Endotelina/genética , Receptores de Endotelina/metabolismo , Sulfonamidas/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA