Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Drug Metab Dispos ; 51(8): 962-969, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37188528

RESUMEN

2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) is an anticancer drug that was discontinued due to hepatotoxicity found in clinical studies. Metabolite analysis of CP-724,714 was conducted using human hepatocytes, in which twelve oxidative metabolites and one hydrolyzed metabolite were formed. Among the three mono-oxidative metabolites, the formation of two was inhibited by adding 1-aminobenzotriazole, a pan-CYP inhibitor. In contrast, the remaining one was not affected by this inhibitor but partially inhibited by hydralazine, indicating that aldehyde oxidase (AO) was involved in metabolizing CP-724,714, which contains a quinazoline substructure, a heterocyclic aromatic quinazoline ring, known to be preferably metabolized by AO. One of the oxidative metabolites of CP-724,714 observed in human hepatocytes was also generated in recombinant human AO. Although CP-724,714 is metabolized by both CYPs and AO in human hepatocytes, the contribution level of AO could not be evaluated using its specific inhibitors because of low AO activity in in vitro human materials. Here, we present a metabolic pathway for CP-724,714 in human hepatocytes and the involvement of AO in CP-724,714 metabolism. We showed here a plausible workflow for predicting AO contribution to the metabolism of CP-724,714 based on DMPK screening data. SIGNIFICANCE STATEMENT: 2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) was identified as a substrate of aldehyde oxidase (AO) rather than xanthine oxidase. Since CP-724,714 is also metabolized by cytochrome P450s (CYPs), the contribution levels of AO and CYPs in the metabolism of CP-724,714 were estimated simultaneously based on in vitro drug metabolism screening data.


Asunto(s)
Aldehído Oxidasa , Sistema Enzimático del Citocromo P-450 , Humanos , Aldehído Oxidasa/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Quinazolinas , Acetamidas
2.
Mol Pharm ; 19(8): 2754-2764, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35766901

RESUMEN

Blood-brain barrier (BBB)-permeable middle- or macromolecules (middle/macromolecules) have recently attracted significant attention as new drug delivery carriers into the human brain via receptor-mediated transcytosis (RMT). During the development process of such carriers, it is necessary to thoroughly evaluate their human BBB permeability levels. In such evaluations, our recently established human immortalized cell-based multicellular spheroidal BBB models (hiMCS-BBB models) have shown high potential. However, the specifics of those capabilities have yet to be elucidated. Therefore, in this study, we characterize the ability of the hiMCS-BBB models to evaluate RMT-mediated BBB penetration properties of middle/macromolecules. More specifically, we began by validating transferrin receptor (TfR)-mediated RMT functionalities using transferrin in the hiMCS-BBB models and then examined the BBB permeability levels of MEM189 antibodies (known BBB-permeable anti-TfR antibodies). The obtained results showed that, as with the case of transferrin, temperature-dependent uptake of MEM189 antibodies was observed in the hiMCS-BBB models, and the extent of that uptake increased in a time-dependent manner until reaching a plateau after around 2 h. To further expand the evaluation applicability of the models, we also examined the BBB permeability levels of the recently developed SLS cyclic peptide and observed that peptide uptake was also temperature-dependent. To summarize, our results show that the hiMCS-BBB models possess the ability to evaluate the RMT-mediated BBB-permeable properties of antibodies and peptides and thus have the potential to provide valuable tools for use in the exploration and identification of middle/macromolecules showing excellent BBB permeability levels, thereby contributing powerfully to the development of new drug delivery carriers for transporting drugs into the human brain.


Asunto(s)
Barrera Hematoencefálica , Receptores de Transferrina , Anticuerpos/química , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Humanos , Receptores de Transferrina/metabolismo , Transcitosis , Transferrina/metabolismo
3.
Pharm Res ; 39(7): 1575-1586, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35288803

RESUMEN

PURPOSE: In vitro human blood-brain barrier (BBB) models in combination with central nervous system-physiologically based pharmacokinetic (CNS-PBPK) modeling, hereafter referred to as the "BBB/PBPK" method, are expected to contribute to prediction of brain drug concentration profiles in humans. As part of our ongoing effort to develop a BBB/PBPK method, we tried to clarify the relationship of in vivo BBB permeability data to those in vitro obtained from a human immortalized cell-based tri-culture BBB model (hiBBB), which we have recently created. METHODS: The hiBBB models were developed and functionally characterized as previously described. The in vitro BBB permeabilities (Pe, × 10-6 cm/s) of seventeen compounds were determined by permeability assays, and in vivo BBB permeabilities (QECF) for eight drugs were estimated by CNS-PBPK modeling. The correlation of the Pe values with the QECF values was analyzed by linear regression analysis. RESULTS: The hiBBB models showed intercellular barrier properties and several BBB transporter functions, which were enough to provide a wide dynamic range of Pe values from 5.7 ± 0.7 (rhodamine 123) to 2580.4 ± 781.9 (rivastigmine). Furthermore, the in vitro Pe values of the eight drugs showed a good correlation (R2 = 0.96) with their in vivo QECF values estimated from human clinical data. CONCLUSION: We show that in vitro human BBB models provide clinically relevant BBB permeability that can be used as input for CNS-PBPK modeling. Therefore, our findings will encourage the development of a BBB/PBPK method as a promising approach for predicting brain drug concentration profiles in humans.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Transporte Biológico/fisiología , Estudios de Factibilidad , Humanos , Permeabilidad
4.
Bioanalysis ; 14(24): 1533-1545, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36825963

RESUMEN

Background: Antibody-drug conjugates (ADCs) are a promising modality for cancer treatment; however, considering their complicated nature, analytical complexity in understanding their pharmacokinetics and pharmacodynamics in the body presents a significant challenge. Results: Vorsetuzumab maleimidocaproyl valine-citrulline p-aminobenzyloxycarbonyl monomethyl auristatin E was used to develop pretreatment and analytical workflows suitable for ADCs. Monomethyl auristatin E release and drug-to-antibody ratio retention were consistent in mouse plasma but inconsistent in monkey and human plasma. Further, metabolites were species-specific. Microflow-liquid chromatography/high-resolution mass spectrometry (LC-HRMS) resulted in a 4-7-fold improvement in detection sensitivity compared with conventional flow LC-HRMS. Conclusion: Microflow-LC-HRMS can be a useful tool in understanding the complex properties of ADCs in the body from a drug metabolism and pharmacokinetics point of view.


Drug-to-antibody ratio (DAR), payload release and metabolite profile of deconjugated payload-linker of vorsetuzumab maleimidocaproyl valine-citrulline p-aminobenzyloxycarbonyl monomethyl auristatin E, an antibody­drug conjugate (ADC) with cleavable linker and monomethyl auristatin E as payload, are reported. Species-specific retention of DAR, payload release and metabolite patterns of deconjugated payload-linker of the ADC are summarized. Exploring the fate of payload-linker moieties deconjugated from ADCs in the body is also vital to understanding pharmacological activity and toxicity. Species-specific metabolite patterns of the ADC provided insight into the importance of optimization of the payload-linker moiety in biological samples, especially in humans. In terms of a more sensitive analytical platform for drug metabolism and pharmacokinetic evaluation, microflow-liquid chromatography/high-resolution mass spectrometry (LC­HRMS) in DAR analysis was found to take advantage of the improvement of detection sensitivity compared with conventional LC­HRMS. Because ADCs are a complex drug modality, these results indicated the importance of evaluation of ADCs from a drug metabolism and pharmacokinetics point of view to understand the pharmacology and toxicology of ADCs, more precisely.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Animales , Ratones , Humanos , Inmunoconjugados/análisis , Espectrometría de Masas/métodos , Cromatografía Liquida/métodos , Haplorrinos/metabolismo
5.
Biol Pharm Bull ; 44(7): 984-991, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33896887

RESUMEN

In vitro blood-brain barrier (BBB) models are essential research tools for use in developing brain-targeted drugs and understanding the physiological and pathophysiological functions of the BBB. To develop BBB models with better functionalities, three-dimensional (3D) culture methods have gained significant attention as a promising approach. In this study, we report on the development of a human conditionally immortalized cell-based multicellular spheroidal BBB (hiMCS-BBB) model. After being seeded into non-attachment culture wells, HASTR/ci35 (astrocytes) and HBPC/ci37 cells (brain pericytes) self-assemble to form a spheroid core that is then covered with an outer monolayer of HBMEC/ci18 cells (brain microvascular endothelial cells). The results of immunocytochemistry showed the protein expression of several cellular junction and BBB-enriched transporter genes in HBMEC/ci18 cells of the spheroid model. The permeability assays showed that the hiMCS-BBB model exhibited barrier functions against the penetration of dextran (5 and 70 kDa) and rhodamine123 (a P-glycoprotein substrate) into the core. On the other hand, facilitation of 2-(N-[7-nitrobenz-2-oxa-1,3-diazol-4-yl]amino)-2-deoxyglucose (2-NBDG; a fluorescent glucose analog) uptake was observed in the hiMCS-BBB model. Furthermore, tumor necrosis factor-alpha treatment elicited an inflammatory response in HBMEC/ci18 cells, thereby suggesting that BBB inflammation can be recapitulated in the hiMCS-BBB model. To summarize, we have developed an hiMCS-BBB model that possesses fundamental BBB properties, which can be expected to provide a useful and highly accessible experimental platform for accelerating various BBB studies.


Asunto(s)
Barrera Hematoencefálica/fisiología , Esferoides Celulares/metabolismo , Astrocitos/metabolismo , Transporte Biológico/fisiología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Humanos , Técnicas In Vitro , Inflamación/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Pericitos/metabolismo , Permeabilidad , Células THP-1 , Factor de Necrosis Tumoral alfa/farmacología
6.
Xenobiotica ; 50(8): 919-928, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32039641

RESUMEN

To reveal putative bioactivation pathways of diclofenac, in vitro human liver materials such as microsomal fractions and hepatocytes were used to confirm metabolic activation of diclofenac by 35S-cysteine trapping assay and covalent binding assay. Candidate human liver proteins possibly targeted by 14C-diclofenac via bioactivation were investigated using two-dimensional gel electrophoresis followed by detection of remaining radioactivity on the modified proteins with bio-imaging analyzer.In the 35S-cysteine trapping assay, three and two adducts with 35S-cysteine were observed in NADPH-fortified and UDPGA-fortified human liver microsomes, respectively. In the covalent binding assay using 14C-diclofenac in human hepatocytes, the extent of covalent binding of diclofenac to human hepatic proteins increased time-dependently. Addition of glutathione attenuated the extent of covalent binding of 14C-diclofenac to human liver microsomal proteins.Fifty-nine proteins from human hepatocytes were proposed as the candidate proteins targeted by reactive metabolites of diclofenac. Proteins modified by cytochrome P450-mediated reactive metabolites were identified by using a cytochrome P450 inhibitor, 1-aminobenzyltriazole and seven of the nine radioactive protein spots were removed by 1-aminobenzyltriazole treatment.In contrast, the remaining two radioactive protein spots, mainly containing human serum albumin and heat shock proteins, were not affected by the addition of 1-aminobenzyltriazole, which suggested the involvement of the acyl glucuronide of diclofenac, formed via uridine diphosphate-glucuronosyl transferases, in the covalent modifications induced by diclofenac.


Asunto(s)
Diclofenaco/metabolismo , Hepatocitos/metabolismo , Antiinflamatorios no Esteroideos/metabolismo , Humanos , Microsomas Hepáticos/metabolismo
8.
Mol Pharm ; 16(11): 4461-4471, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31573814

RESUMEN

Brain microvascular endothelial cells (BMEC), together with astrocytes and pericytes, form the blood-brain barrier (BBB) that strictly restricts drug penetration into the brain. Therefore, in central nervous system drug development, the establishment of an in vitro human BBB model for use in studies estimating the in vivo human BBB permeability of drug candidates has long been awaited. The current study developed and characterized a human immortalized cell-based BBB triculture model, termed the "hiBBB" model. To set up the hiBBB model, human immortalized BMEC (HBMEC/ci18) were cocultured with human immortalized astrocytes (HASTR/ci35) and brain pericytes (HBPC/ci37) in a transwell system. The trans-endothelial electrical resistance of the hiBBB model was 134.4 ± 5.5 (Ω × cm2), and the efflux ratios of rhodamine123 and dantrolene were 1.72 ± 0.11 and 1.72 ± 0.45, respectively, suggesting that the hiBBB model possesses essential cellular junction and efflux transporter functions. In BBB permeability assays, the mean value of the permeability coefficients (Pe) of BBB permeable compounds (propranolol, pyrilamine, memantine, and diphenhydramine) was 960 × 10-6 cm/s, which was clearly distinguishable from that of BBB nonpermeable compounds (sodium fluorescein and Lucifer yellow, 18 × 10-6 cm/s). Collectively, this study successfully developed the hiBBB model, which exhibits essential BBB functionality. Taking into consideration the high availability of the immortalized cells used in the hiBBB model, our results are expected to become an initial step toward the establishment of a useful human BBB model to investigate drug penetration into the human brain.


Asunto(s)
Transporte Biológico/fisiología , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Preparaciones Farmacéuticas/metabolismo , Astrocitos/metabolismo , Línea Celular , Técnicas de Cocultivo/métodos , Células Endoteliales/metabolismo , Humanos , Pericitos/metabolismo , Permeabilidad
9.
Mol Pharm ; 15(6): 2277-2288, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29746136

RESUMEN

In vitro-in vivo extrapolation based on uptake clearance determined in human hepatocytes has been used to predict in vivo hepatic clearance of organic anion transporting polypeptide (OATP) substrates. This study evaluated the relative activity factor (RAF) approach to extrapolate active uptake clearance in transporter-transfected cell systems (CLuptake) to that in human hepatocyte suspensions (PSinf,act). RAF values for OATP1B1 and OATP1B3 were determined in two batches of cryopreserved human hepatocytes using estrone-3-sulfate and cholecystokinin octapeptide as reference substrates, respectively. Fourteen OATP1B substrate drugs selected (atorvastatin, bosentan, cerivastatin, fexofenadine, fluvastatin, glibenclamide, irbesartan, nateglinide, pitavastatin, pravastatin, rosuvastatin, telmisartan, torasemide, and valsartan) showed temperature-dependent uptake in human hepatocytes. In transporter-transfected cells, OATP1B1- and OATP1B3-mediated uptake was observed in all compounds except for telmisartan. RAF-based net CLuptake was mainly accounted for by OATP1B1 (72.3-99.7%) and fell within the 3-fold of PSinf,act observed in human hepatocytes in 11 out of 13 compounds (excluding telmisartan). This study demonstrated that the RAF approach provides a quantitative index of OATP1B1- and OATP1B3-mediated PSinf,act in human hepatocytes, which will facilitate the optimization of the pharmacokinetic properties of OATP1B substrates at nonclinical stages of drug development.


Asunto(s)
Hepatocitos/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Preparaciones Farmacéuticas/metabolismo , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/metabolismo , Colecistoquinina/metabolismo , Estrona/análogos & derivados , Estrona/metabolismo , Células HEK293 , Humanos
10.
J Pharm Sci ; 106(9): 2678-2687, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28238896

RESUMEN

Prediction of human pharmacokinetic profiles of drug candidates is an essential step toward first-in-human studies. However, it remains difficult to quantitatively predict hepatic clearance, particularly when hepatic uptake is mediated by transporter(s). Using 15 organic anion transporting polypeptide (OATP) substrate drugs, we tested 3 in vitro-in vivo extrapolation (IVIVE) approaches to predict overall hepatic intrinsic clearance in vivo (CLint,all,vivo). IVIVE approaches involved metabolic intrinsic clearance in human liver microsomes (CLint,met) with or without hepatocyte-to-buffer maximum unbound concentration ratio (Kp,uu,max) correction and uptake intrinsic clearance at 37°C (PSinf,37°C) in human hepatocyte suspensions. Kp,uu,max and PSinf,37°C values were determined in 2 hepatocyte batches, and all tested compounds showed temperature-dependent uptake, consistent with the fact of transporter-mediated uptake. CLint,met substantially underestimated CLint,all,vivo. By multiplying CLint,met by Kp,uu,max values, the prediction performance was much improved; however, in vitro-in vivo correlation was poor. Among the IVIVE approaches, PSinf,37°C showed the most robust correlation with CLint,all,vivo, and one of the hepatocyte batches could predict CLint,all,vivo with a minimal empirical scaling factor. These results suggested IVIVE with hepatic uptake clearance determined in hepatocyte suspensions as one of the most practical approaches for predicting CLint,all,vivo of OATP substrate drugs.


Asunto(s)
Hígado/metabolismo , Microsomas Hepáticos/metabolismo , Modelos Biológicos , Transportadores de Anión Orgánico/metabolismo , Transporte Biológico , Glucuronosiltransferasa/metabolismo , Hepatocitos , Humanos , Cinética , Tasa de Depuración Metabólica , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Espectrometría de Masas en Tándem/métodos , Temperatura
11.
Epilepsia ; 57(4): 638-47, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26854031

RESUMEN

OBJECTIVE: Anticonvulsive monotherapy fails to be effective in one third of patients with epilepsy resulting in the need for polytherapy regimens. However, with the still limited knowledge, drug choices for polytherapy remain empirical. Here we report experimental data from a chronic epilepsy model for the combination of perampanel and zonisamide, which can render guidance for clinical studies and individual drug choices. METHODS: The anticonvulsant effects of the combination of perampanel and zonisamide were evaluated in a rat amygdala kindling model. Furthermore, the potential for motor impairment was evaluated. The type of interaction was quantitatively assessed based on isobolographic analysis. RESULTS: When administered alone, zonisamide dose-dependently increased the afterdischarge threshold in fully kindled rats. Moreover, data confirmed efficacy of perampanel to inhibit seizure initiation and progression with an impact on propagation of activity from the focus. Pronounced threshold increases were observed following administration of a constant zonisamide dosage combined with different doses of perampanel. Isobolographic analysis of drug responses, which is based on individual drug dose-effect data, revealed a synergistic interaction substantiating the high efficacy of the combination. Furthermore, rotarod data indicated that the combination has a favorable tolerability profile when zonisamide is coadministered with low doses of perampanel. Plasma concentration analysis argued against a pharmacokinetic interaction as a basis for the synergism. SIGNIFICANCE: The findings clearly indicate a pronounced synergistic anticonvulsant effect for the combination of the noncompetitive α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist perampanel with zonisamide, which modulates voltage-sensitive sodium channels and T-type calcium currents. Consequently, polytherapy using these two antiepileptic drugs might be efficacious for clinical management of partial-onset seizures. The findings indicate that the impact of dose ratios on tolerability needs be taken into account. With regard to conclusions about the extent of the synergism and its implications further antiepileptic drug combinations need to be evaluated allowing direct comparison.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Isoxazoles/administración & dosificación , Excitación Neurológica/efectos de los fármacos , Piridonas/administración & dosificación , Animales , Anticonvulsivantes/sangre , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Epilepsia del Lóbulo Temporal/sangre , Femenino , Isoxazoles/sangre , Excitación Neurológica/metabolismo , Nitrilos , Piridonas/sangre , Ratas , Ratas Sprague-Dawley , Zonisamida
12.
Mol Pharm ; 13(2): 438-48, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26696140

RESUMEN

Organic anion transporting polypeptide (OATP) 1B1 plays an important role in the hepatic uptake of various drugs. Because OATP1B1 is a site of drug-drug interactions (DDIs), evaluating the inhibitory potential of drug candidates on OATP1B1 is required during drug development. For establishing a highly sensitive, high-throughput fluorescence-based OATP1B1 inhibition assay system, the present study focused on fluorescein (FL) and its derivatives and evaluated their uptake via OATP1B1 as well as OATP1B3 and OATP2B1 using the transporter-expressing human embryonic kidney 293 cells. We identified 2',7'-dichlorofluorescein (DCF), 4',5'-dibromofluorescein (DBF), and Oregon green (OG) as good OATP1B1 substrates with Km values of 5.29, 4.16, and 54.1 µM and Vmax values of 87.9, 48.1, and 187 pmol/min/mg protein, respectively. In addition to FL, fluo-3, and 8-fluorescein-cAMP, OG, and DBF were identified as OATP1B3 substrates. FL, OG, DCF, and DBF were identified as OATP2B1 substrates. Among the FL derivatives, DCF displayed the highest OATP1B1-mediated uptake. The Ki values of 14 compounds on OATP1B1 determined with DCF as a probe exhibited good agreement with those obtained using [(3)H]estradiol-17ß-glucuronide (E2G) as a substrate, whereas [(3)H]estrone-3-sulfate and [(3)H]sulfobromophthalein yielded higher Ki values for all inhibitors than DCF. Mutually competitive inhibition observed between DCF and E2G suggested that they share the same binding site on OATP1B1. Therefore, DCF as well as E2G can be used as sensitive probes for in vitro OATP1B1 inhibition assays, which will help mitigate the risk of false-negative DDI predictions potentially caused by substrate-dependent Ki variations.


Asunto(s)
Bioensayo/métodos , Fluoresceína/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado/antagonistas & inhibidores , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Transporte Biológico , Fluoresceína/química , Fluorescencia , Células HEK293 , Humanos
13.
Drug Metab Dispos ; 43(2): 235-47, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25414411

RESUMEN

The risk assessment of organic anion transporting polypeptide (OATP) 1B1-mediated drug-drug interactions (DDIs) is an indispensable part of drug development. We previously reported that in vitro inhibitory potencies of several inhibitors on OATP1B1 depend on the substrates when prototypical substrates, estradiol-17ß-glucuronide (E2G), estrone-3-sulfate, and sulfobromophthalein were used as test substrates. The purpose of this study was to comprehensively investigate this substrate-dependent inhibition of OATP1B1 using clinically relevant OATP1B1 inhibitors and substrate drugs. Effects of cyclosporine A (CsA), rifampin, and gemfibrozil on OATP1B1-mediated uptake of 12 substrate drugs were examined in OATP1B1-expressing human embryonic kidney 293 cells. The Ki values (µM) for CsA varied from 0.0771 to 0.486 (6.3-fold), for rifampin from 0.358 to 1.23 (3.4-fold), and for gemfibrozil from 9.65 to 252 (26-fold). Except for the inhibition of torasemide uptake by CsA and that of nateglinide uptake by gemfibrozil, the Ki values were within 2.8-fold of those obtained using E2G as a substrate. Preincubation potentiated the inhibitory effect of CsA on OATP1B1 with similar magnitude regardless of the substrates. R values calculated based on a static model showed some variation depending on the Ki values determined with various substrates, and such variability could have an impact on the DDI predictions particularly for a weak-to-moderate inhibitor (gemfibrozil). OATP1B1 substrate drugs except for torasemide and nateglinide, or E2G as a surrogate, is recommended as an in vitro probe in the inhibition experiments, which will help mitigate the risk of false-negative DDI predictions potentially caused by substrate-dependent Ki variation.


Asunto(s)
Antihipertensivos/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/metabolismo , Hipoglucemiantes/metabolismo , Moduladores del Transporte de Membrana/farmacología , Modelos Biológicos , Transportadores de Anión Orgánico/antagonistas & inhibidores , Unión Competitiva , Transporte Biológico/efectos de los fármacos , Ciclohexanos/metabolismo , Ciclosporina/farmacología , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Estradiol/análogos & derivados , Estradiol/metabolismo , Gemfibrozilo/farmacología , Células HEK293 , Humanos , Cinética , Transportador 1 de Anión Orgánico Específico del Hígado , Nateglinida , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Fenilalanina/análogos & derivados , Fenilalanina/metabolismo , Proteínas Recombinantes/metabolismo , Rifampin/farmacología , Sulfonamidas/metabolismo , Torasemida
14.
Drug Metab Dispos ; 41(10): 1859-66, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23920221

RESUMEN

Organic anion transporting polypeptide (OATP) 1B1 plays an important role in the hepatic uptake of many drugs, and the evaluation of OATP1B1-mediated drug-drug interactions (DDIs) is emphasized in the latest DDI (draft) guidance documents from U.S. and E.U. regulatory agencies. It has been suggested that some OATP1B1 inhibitors show a discrepancy in their inhibitory potential, depending on the substrates used in the cell-based assay. In this study, inhibitory effects of 14 compounds on the OATP1B1-mediated uptake of the prototypical substrates [³H]estradiol-17ß-glucuronide (E2G), [³H]estrone-3-sulfate (E1S), and [³H]sulfobromophthalein (BSP) were studied in OATP1B1-transfected cells. Inhibitory potencies of tested compounds varied depending on the substrates. Ritonavir, gemfibrozil, and erythromycin caused remarkable substrate-dependent inhibition with up to 117-, 14-, and 13-fold difference in their IC50 values, respectively. Also, the clinically relevant OATP inhibitors rifampin and cyclosporin A exhibited up to 12- and 6-fold variation in their IC50 values, respectively. Regardless of the inhibitors tested, the most potent OATP1B1 inhibition was observed when [³H]E2G was used as a substrate. Mutual inhibition studies of OATP1B1 indicated that E2G and E1S competitively inhibited each other, whereas BSP noncompetitively inhibited E2G uptake. In addition, BSP inhibited E1S in a competitive manner, but E1S caused an atypical kinetics on BSP uptake. This study showed substrate-dependent inhibition of OATP1B1 and demonstrated that E2G was the most sensitive in vitro OATP1B1 probe substrate among three substrates tested. This will give us an insight into the assessment of clinically relevant OATP1B1-mediated DDI in vitro with minimum potential of false-negative prediction.


Asunto(s)
Estradiol/análogos & derivados , Estrona/análogos & derivados , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Transportadores de Anión Orgánico/antagonistas & inhibidores , Sulfobromoftaleína/metabolismo , Transporte Biológico/efectos de los fármacos , Línea Celular , Estradiol/metabolismo , Estrona/metabolismo , Células HEK293 , Humanos , Cinética , Transportador 1 de Anión Orgánico Específico del Hígado , Transportadores de Anión Orgánico/metabolismo
15.
Am J Physiol Renal Physiol ; 299(4): F767-75, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668102

RESUMEN

Apical reabsorption from the urine has been shown to be important for such processes as the maintenance of critical metabolites in the blood and the excretion of nephrotoxic compounds. The solute carrier (SLC) transporter OAT4 (SLC22A11) is expressed on the apical membrane of renal proximal tubule cells and is known to mediate the transport of a variety of xenobiotic and endogenous organic anions. Functional characterization of genetic variants of apical transporters thought to mediate reabsorption, such as OAT4, may provide insight into the genetic factors influencing the complex pathways involved in drug elimination and metabolite reclamation occurring in the kidney. Naturally occurring genetic variants of OAT4 were identified in public databases and by resequencing DNA samples from 272 individuals comprising 4 distinct ethnic groups. Nine total nonsynonymous variants were identified and functionally assessed using uptake of three radiolabeled substrates. A nonsense variant, R48Stop, and three other variants (R121C, V155G, and V155M) were found at frequencies of at least 2% in an ethnic group specific fashion. The L29P, R48Stop, and H469R variants displayed a complete loss of function, and kinetic analysis identified a reduced V(max) in the common nonsynonymous variants. Plasma membrane levels of OAT4 protein were absent or reduced in the nonfunctional variants, providing a mechanistic reason for the observed loss of function. Characterization of the genetic variants of reabsorptive transporters such as OAT4 is an important step in understanding variability in tubular reabsorption with important implications in innate homeostatic processes and drug disposition.


Asunto(s)
ADN/genética , Variación Genética/fisiología , Riñón/fisiología , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/fisiología , Negro o Afroamericano/genética , Secuencia de Aminoácidos , Asiático/genética , Transporte Biológico/fisiología , Células Cultivadas , Humanos , Americanos Mexicanos/genética , Datos de Secuencia Molecular , Transfección , Población Blanca/genética
16.
Mol Pharmacol ; 73(4): 1151-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18216183

RESUMEN

The second messenger, cGMP, mediates a host of cellular responses to various stimuli, resulting in the regulation of many critical physiologic functions. The existence of specific cGMP transporters on the plasma membrane that participate in the regulation of cGMP levels has been suggested in a large number of studies. In this study, we identified a novel plasma membrane transporter for cGMP. In particular, we showed that hOAT2 (SLC22A7), a member of the solute carrier (SLC) superfamily, was a facilitative transporter for cGMP and other guanine nucleotides. hOAT2, which is ubiquitously expressed at high levels in many cell types, was previously thought to primarily transport organic anions. Among purine and pyrimidine nucleobases, nucleosides, and nucleotides, hOAT2 showed the greatest preference for cGMP, which transported cGMP with a K(m) value of 88 +/- 11 muM and exhibited between 50- and 100-fold enhanced uptake over control cells. Our data revealed that hOAT2 is a bidirectional facilitative transporter that can control both intracellular and extracellular levels of cGMP. In addition, we observed that a common alternatively spliced variant of hOAT2 demonstrated a complete loss of transport function as a result of a low expression level on the plasma membrane. We conclude that hOAT2 is a highly efficient, facilitative transporter of cGMP and may be involved in cGMP signaling in many tissues. Our study suggests that hOAT2 represents a potential new drug target for regulating cGMP levels.


Asunto(s)
GMP Cíclico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Empalme Alternativo/genética , Aminoácidos , Transporte Biológico , Línea Celular , Desoxiguanosina/metabolismo , Espacio Extracelular/metabolismo , Perfilación de la Expresión Génica , Humanos , Espacio Intracelular/metabolismo , Nucleósidos/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/química , Transportadores de Anión Orgánico Sodio-Independiente/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especificidad por Sustrato , Transfección
17.
Cancer Res ; 66(17): 8847-57, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16951202

RESUMEN

Although the platinum-based anticancer drugs cisplatin, carboplatin, and oxaliplatin have similar DNA-binding properties, only oxaliplatin is active against colorectal tumors. The mechanisms for this tumor specificity of platinum-based compounds are poorly understood but could be related to differences in uptake. This study shows that the human organic cation transporters (OCT) 1 and 2 (SLC22A1 and SLC22A2) markedly increase oxaliplatin, but not cisplatin or carboplatin, accumulation and cytotoxicity in transfected cells, indicating that oxaliplatin is an excellent substrate of these transporters. The cytotoxicity of oxaliplatin was greater than that of cisplatin in six colon cancer cell lines [mean +/- SE of IC(50) in the six cell lines, 3.9 +/- 1.4 micromol/L (oxaliplatin) versus 11 +/- 2.0 micromol/L (cisplatin)] but was reduced by an OCT inhibitor, cimetidine, to a level similar to, or even lower than that of, cisplatin (29 +/- 11 micromol/L for oxaliplatin versus 19 +/- 4.3 micromol/L for cisplatin). Structure-activity studies indicated that organic functionalities on nonleaving groups coordinated to platinum are critical for selective uptake by OCTs. These results indicate that OCT1 and OCT2 are major determinants of the anticancer activity of oxaliplatin and may contribute to its antitumor specificity. They also strongly suggest that expression of OCTs in tumors should be investigated as markers for selecting specific platinum-based therapies in individual patients. The development of new anticancer drugs, specifically targeted to OCTs, represents a novel strategy for targeted drug therapy. The results of the present structure-activity studies indicate specific tactics for realizing this goal.


Asunto(s)
Antineoplásicos/toxicidad , Factor 1 de Transcripción de Unión a Octámeros/genética , Proteínas de Transporte de Catión Orgánico/genética , Compuestos Organoplatinos/toxicidad , Piridinas/toxicidad , Animales , Línea Celular , Cisplatino/toxicidad , Aductos de ADN , Perros , Vectores Genéticos , Humanos , Riñón , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Platino (Metal) , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Relación Estructura-Actividad , Transfección
18.
Chirality ; 14(7): 599-603, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12112335

RESUMEN

The investigation was undertaken to study the stereoselective protein binding of alprenolol in renal disease patient sera, compared to that in the sera of healthy volunteers. The in vitro stereoselective protein binding of beta-blockers was determined in undiluted serum and in isolated alpha(1)-acid glycoprotein (AGP) solutions by ultrafiltration. The stereoselctive serum protein binding of alprenolol, a beta-adrenergic blocking agent, in healthy volunteers was significantly altered in renal disease patients. We investigated the effects of AGP concentration and endogenous substances, including uremic toxins, on the stereoselective protein binding of alprenolol in renal disease patients. A good correlation between the unbound (R)/(S) ratio (F(R)/F(S) ratio), an apparent index of stereoselectivity in alprenolol serum binding and AGP concentration in serum, was found. However, stereoselective protein binding was not influenced by endogenous substances. This result can be explained by the difference in binding affinities of (R) and (S)-isomers of alprenolol to AGP. We conclude that the stereoselective protein binding of alprenolol in healthy volunteers and renal disease patients varies as a result of changes in AGP concentration. Accordingly, these findings might be useful in alprenolol therapy in renal disease patients.


Asunto(s)
Alprenolol/química , Enfermedades Renales/sangre , Orosomucoide/química , Alprenolol/sangre , Antihipertensivos/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Enfermedades Renales/terapia , Orosomucoide/aislamiento & purificación , Unión Proteica , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...