51
|
Ufuk A, Kosa RE, Gao H, Bi YA, Modi S, Gates D, Rodrigues AD, Tremaine LM, Varma MVS, Houston JB, Galetin A. In Vitro-In Vivo Extrapolation of OATP1B-Mediated Drug-Drug Interactions in Cynomolgus Monkey. J Pharmacol Exp Ther 2018; 365:688-699. [PMID: 29643253 DOI: 10.1124/jpet.118.247767] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/06/2018] [Indexed: 12/31/2022] Open
Abstract
Hepatic organic anion-transporting polypeptides (OATP) 1B1 and 1B3 are clinically relevant transporters associated with significant drug-drug interactions (DDIs) and safety concerns. Given that OATP1Bs in cynomolgus monkey share >90% degree of gene and amino acid sequence homology with human orthologs, we evaluated the in vitro-in vivo translation of OATP1B-mediated DDI risk using this preclinical model. In vitro studies using plated cynomolgus monkey hepatocytes showed active uptake Km values of 2.0 and 3.9 µM for OATP1B probe substrates, pitavastatin and rosuvastatin, respectively. Rifampicin inhibited pitavastatin and rosuvastatin active uptake in monkey hepatocytes with IC50 values of 3.0 and 0.54 µM, respectively, following preincubation with the inhibitor. Intravenous pharmacokinetics of 2H4-pitavastatin and 2H6-rosuvastatin (0.2 mg/kg) and the oral pharmacokinetics of cold probes (2 mg/kg) were studied in cynomolgus monkeys (n = 4) without or with coadministration of single oral ascending doses of rifampicin (1, 3, 10, and 30 mg/kg). A rifampicin dose-dependent reduction in i.v. clearance of statins was observed. Additionally, oral pitavastatin and rosuvastatin plasma exposure increased up to 19- and 15-fold at the highest dose of rifampicin, respectively. Use of in vitro IC50 obtained following 1 hour preincubation with rifampicin (0.54 µM) predicted correctly the change in mean i.v. clearance and oral exposure of statins as a function of mean unbound maximum plasma concentration of rifampicin. This study demonstrates quantitative translation of in vitro OATP1B IC50 to predict DDIs using cynomolgus monkey as a preclinical model and provides further confidence in application of in vitro hepatocyte data for the prediction of clinical OATP1B-mediated DDIs.
Collapse
Affiliation(s)
- Ayşe Ufuk
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Rachel E Kosa
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Hongying Gao
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Yi-An Bi
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Sweta Modi
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Dana Gates
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - A David Rodrigues
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Larry M Tremaine
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Manthena V S Varma
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (A.U., J.B.H., A.G.); and Pharmacokinetics, Dynamics, and Metabolism (R.E.K., H.G., Y.-A.B., A.D.R., L.M.T., M.V.S.V.) and Research Formulations, Pharmaceutical Sciences (S.M., D.G.), Medicine Design, Pfizer Worldwide R&D, Groton, Connecticut
| |
Collapse
|
52
|
Izumi S, Nozaki Y, Kusuhara H, Hotta K, Mochizuki T, Komori T, Maeda K, Sugiyama Y. Relative Activity Factor (RAF)-Based Scaling of Uptake Clearance Mediated by Organic Anion Transporting Polypeptide (OATP) 1B1 and OATP1B3 in Human Hepatocytes. Mol Pharm 2018; 15:2277-2288. [DOI: 10.1021/acs.molpharmaceut.8b00138] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Saki Izumi
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yoshitane Nozaki
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical
Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-003, Japan
| | - Koichiro Hotta
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Toshiki Mochizuki
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Takafumi Komori
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical
Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-003, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama-shi, Kanagawa 230-0045, Japan
| |
Collapse
|
53
|
Identification of novel cell-impermeant fluorescent substrates for testing the function and drug interaction of Organic Anion-Transporting Polypeptides, OATP1B1/1B3 and 2B1. Sci Rep 2018; 8:2630. [PMID: 29422623 PMCID: PMC5805760 DOI: 10.1038/s41598-018-20815-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/19/2018] [Indexed: 12/15/2022] Open
Abstract
Organic Anion-Transporting Polypeptides are multispecific membrane proteins that regulate the passage of crucial endobiotics and drugs across pharmacological barriers. OATP1B1 and OATP1B3 have been described to play a major role in the hepatic uptake of statins, antivirals and various chemotherapeutics; whereas the pharmacological role of the ubiquitously expressed OATP2B1 is less well characterized. According to current industry standards, in vitro testing for susceptibility to OATP1B1 and 1B3 mediated transport is recommended for drug candidates that are eliminated in part via the liver. Here we show that human OATP1B1, 1B3 and 2B1 transport a series of commercially available viability dyes that are generally believed to be impermeable to intact cells. We demonstrate that the intracellular accumulation of Zombie Violet, Live/Dead Green, Cascade Blue and Alexa Fluor 405 is specifically increased by OATPs. Inhibition of Cascade Blue or Alexa Fluor 405 uptake by known OATP substrates/inhibitors yielded IC50 values in agreement with gold-standard radioligand assays. The fluorescence-based assays described in this study provide a new tool for testing OATP1B/2B1 drug interactions.
Collapse
|
54
|
Koide H, Tsujimoto M, Takeuchi A, Tanaka M, Ikegami Y, Tagami M, Abe S, Hashimoto M, Minegaki T, Nishiguchi K. Substrate-dependent effects of molecular-targeted anticancer agents on activity of organic anion transporting polypeptide 1B1. Xenobiotica 2017; 48:1059-1071. [DOI: 10.1080/00498254.2017.1393582] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Hiroyoshi Koide
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Masayuki Tsujimoto
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ai Takeuchi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Miyu Tanaka
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yoko Ikegami
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Mayu Tagami
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Syoko Abe
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Miki Hashimoto
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tetsuya Minegaki
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kohshi Nishiguchi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
55
|
Mathialagan S, Costales C, Tylaska L, Kimoto E, Vildhede A, Johnson J, Johnson N, Sarashina T, Hashizume K, Isringhausen CD, Vermeer LMM, Wolff AR, Rodrigues AD. In vitro studies with two human organic anion transporters: OAT2 and OAT7. Xenobiotica 2017; 48:1037-1049. [DOI: 10.1080/00498254.2017.1384595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sumathy Mathialagan
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Chester Costales
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Laurie Tylaska
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Emi Kimoto
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Anna Vildhede
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Jillian Johnson
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Nathaniel Johnson
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | | | | | | | | | | | - A. David Rodrigues
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| |
Collapse
|
56
|
Shen H, Chen W, Drexler DM, Mandlekar S, Holenarsipur VK, Shields EE, Langish R, Sidik K, Gan J, Humphreys WG, Marathe P, Lai Y. Comparative Evaluation of Plasma Bile Acids, Dehydroepiandrosterone Sulfate, Hexadecanedioate, and Tetradecanedioate with Coproporphyrins I and III as Markers of OATP Inhibition in Healthy Subjects. Drug Metab Dispos 2017; 45:908-919. [PMID: 28576766 DOI: 10.1124/dmd.117.075531] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/31/2017] [Indexed: 12/20/2022] Open
Abstract
Multiple endogenous compounds have been proposed as candidate biomarkers to monitor organic anion transporting polypeptide (OATP) function in preclinical species or humans. Previously, we demonstrated that coproporphyrins (CPs) I and III are appropriate clinical markers to evaluate OATP inhibition and recapitulate clinical drug-drug interactions (DDIs). In the present study, we investigated bile acids (BAs) dehydroepiandrosterone sulfate (DHEAS), hexadecanedioate (HDA), and tetradecanedioate (TDA) in plasma as endogenous probes for OATP inhibition and compared these candidate probes to CPs. All probes were determined in samples from a single study that examined their behavior and their association with rosuvastatin (RSV) pharmacokinetics after administration of an OATP inhibitor rifampin (RIF) in healthy subjects. Among endogenous probes examined, RIF significantly increased maximum plasma concentration (Cmax) and area under the concentration-time curve (AUC)(0-24h) of fatty acids HDA and TDA by 2.2- to 3.2-fold. For the 13 bile acids in plasma examined, no statistically significant changes were detected between treatments. Changes in plasma DHEAS did not correlate with OATP1B inhibition by RIF. On the basis of the magnitude of effects for the endogenous compounds that demonstrated significant changes from baseline over interindividual variations, the overall rank order for the AUC change was found to be CP I > CP III > HDA ≈ TDA ≈ RSV > > BAs. Collectively, these results reconfirmed that CPs are novel biomarkers suitable for clinical use. In addition, HDA and TDA are useful for OATP functional assessment. Since these endogenous markers can be monitored in conjunction with pharmacokinetics analysis, the CPs and fatty acid dicarboxylates, either alone or in combination, offer promise of earlier diagnosis and risk stratification for OATP-mediated DDIs.
Collapse
Affiliation(s)
- Hong Shen
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Weiqi Chen
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Dieter M Drexler
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Sandhya Mandlekar
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Vinay K Holenarsipur
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Eric E Shields
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Robert Langish
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Kurex Sidik
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Jinping Gan
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - W Griffith Humphreys
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Punit Marathe
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| | - Yurong Lai
- Pharmaceutical Candidate Optimization (H.S., W.C., R.L., J.G., W.G.H., P.M., Y.L.) and Global Biometrics Sciences (K.S.), Bristol-Myers Squibb Company, Princeton, New Jersey; Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Wallingford, Connecticut (D.M.D., E.E.S.); Bristol-Myers Squibb India Pvt. Ltd. (S.M.) and Syngene International Ltd. (V.K.H.), Biocon BMS R&D Center, Bangalore, India
| |
Collapse
|
57
|
Pahwa S, Alam K, Crowe A, Farasyn T, Neuhoff S, Hatley O, Ding K, Yue W. Pretreatment With Rifampicin and Tyrosine Kinase Inhibitor Dasatinib Potentiates the Inhibitory Effects Toward OATP1B1- and OATP1B3-Mediated Transport. J Pharm Sci 2017; 106:2123-2135. [PMID: 28373111 PMCID: PMC5511785 DOI: 10.1016/j.xphs.2017.03.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 03/08/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
Present studies determined the effects of pretreatment with rifampicin, an organic anion-transporting polypeptide (OATP) inhibitor, and the tyrosine kinase inhibitor dasatinib on OATP1B1- and OATP1B3-mediated transport, and evaluated the OATP-mediated drug-drug interaction potential of dasatinib using the static R-value and dynamic physiologically based pharmacokinetic models. Rifampicin and dasatinib pretreatment significantly decreased OATP1B1- and OATP1B3-mediated transport. Rifampicin pretreatment also significantly decreased [3H]-pitavastatin and [3H]-CCK-8 accumulation in human sandwich-cultured hepatocytes. Present studies revealed that estrone-3-sulfate is a less-sensitive OATP1B1 substrate than estradiol-17β-glucuronide in assessing rifampicin pretreatment effects. Pretreatment with rifampicin and dasatinib reduced the inhibition constant (Ki) values against OATP1B1 by 3 and 2.1 fold and against OATP1B3 by 2.4 and 2.1 fold, respectively. The in vitro rifampicin Ki values after preincubation are comparable to the estimated in vivo Ki reported previously. Models predict that dasatinib has a low potential to cause OATP1B1- and OATP1B3-mediated drug-drug interactions. Time-lapse confocal microscopy demonstrated that rifampicin and dasatinib pretreatment did not affect plasma membrane localization of green-fluorescent protein-tagged OATP1B1 (GFP-OATP1B1) and GFP-OATP1B3 in human embryonic kidney 293 stable cell lines. In summary, we report novel findings that pretreatment with rifampicin and dasatinib potentiates the inhibitory effects toward OATP1B1 and OATP1B3 without affecting plasma membrane levels of the transporters.
Collapse
Affiliation(s)
- Sonia Pahwa
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117
| | - Khondoker Alam
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117
| | - Alexandra Crowe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117
| | - Taleah Farasyn
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117
| | - Sibylle Neuhoff
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield S2 4SU, UK
| | - Oliver Hatley
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield S2 4SU, UK
| | - Kai Ding
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117.
| |
Collapse
|
58
|
Lee SC, Arya V, Yang X, Volpe DA, Zhang L. Evaluation of transporters in drug development: Current status and contemporary issues. Adv Drug Deliv Rev 2017; 116:100-118. [PMID: 28760687 DOI: 10.1016/j.addr.2017.07.020] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 01/22/2023]
Abstract
Transporters govern the access of molecules to cells or their exit from cells, thereby controlling the overall distribution of drugs to their intracellular site of action. Clinically relevant drug-drug interactions mediated by transporters are of increasing interest in drug development. Drug transporters, acting alone or in concert with drug metabolizing enzymes, can play an important role in modulating drug absorption, distribution, metabolism and excretion, thus affecting the pharmacokinetics and/or pharmacodynamics of a drug. The drug interaction guidance documents from regulatory agencies include various decision criteria that may be used to predict the need for in vivo assessment of transporter-mediated drug-drug interactions. Regulatory science research continues to assess the prediction performances of various criteria as well as to examine the strength and limitations of each prediction criterion to foster discussions related to harmonized decision criteria that may be used to facilitate global drug development. This review discusses the role of transporters in drug development with a focus on methodologies in assessing transporter-mediated drug-drug interactions, challenges in both in vitro and in vivo assessments of transporters, and emerging transporter research areas including biomarkers, assessment of tissue concentrations, and effect of diseases on transporters.
Collapse
Affiliation(s)
- Sue-Chih Lee
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Vikram Arya
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Xinning Yang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Donna A Volpe
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
59
|
Takehara I, Terashima H, Nakayama T, Yoshikado T, Yoshida M, Furihata K, Watanabe N, Maeda K, Ando O, Sugiyama Y, Kusuhara H. Investigation of Glycochenodeoxycholate Sulfate and Chenodeoxycholate Glucuronide as Surrogate Endogenous Probes for Drug Interaction Studies of OATP1B1 and OATP1B3 in Healthy Japanese Volunteers. Pharm Res 2017; 34:1601-1614. [PMID: 28550384 DOI: 10.1007/s11095-017-2184-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 05/15/2017] [Indexed: 01/26/2023]
Abstract
PURPOSE To assess the use of glycochenodeoxycholate-3-sulfate (GCDCA-S) and chenodeoxycholate 3- or 24-glucuronide (CDCA-3G or -24G) as surrogate endogenous substrates in the investigation of drug interactions involving OATP1B1 and OATP1B3. METHODS Uptake of GCDCA-S and CDCA-24G was examined in HEK293 cells transfected with cDNA for OATP1B1, OATP1B3, and NTCP and in cryopreserved human hepatocytes. Plasma concentrations of bile acids and their metabolites (GCDCA-S, CDCA-3G, and CDCA-24G) were determined by LC-MS/MS in eight healthy volunteers with or without administration of rifampicin (600 mg, po). RESULTS GCDCA-S and CDCA-24G were substrates for OATP1B1, OATP1B3, and NTCP. The uptake of [3H]atorvastatin, GCDCA-S, and CDCA-24G by human hepatocytes was significantly inhibited by both rifampicin and pioglitazone, whereas that of taurocholate was inhibited only by pioglitazone. Rifampicin elevated plasma concentrations of GCDCA-S more than those of other bile acids. The area under the plasma concentration-time curve for GCDCA-S was 20.3 times higher in rifampicin-treated samples. CDCA-24G could be detected only in plasma from the rifampicin-treatment phase, and CDCA-3G was undetectable in both phases. CONCLUSIONS We identified GCDCA-S and CDCA-24G as substrates of NTCP, OATP1B1, and OATP1B3. GCDCA-S is a surrogate endogenous probe for the assessment of drug interactions involving hepatic OATP1B1 and OATP1B3.
Collapse
Affiliation(s)
- Issey Takehara
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Biomarker Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Hanano Terashima
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takeshi Nakayama
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takashi Yoshikado
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Japan
| | - Miwa Yoshida
- P-One Clinic, Keikokai Medical Corp, Tokyo, Japan
| | | | - Nobuaki Watanabe
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Osamu Ando
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
60
|
Bins S, van Doorn L, Phelps MA, Gibson AA, Hu S, Li L, Vasilyeva A, Du G, Hamberg P, Eskens F, de Bruijn P, Sparreboom A, Mathijssen R, Baker SD. Influence of OATP1B1 Function on the Disposition of Sorafenib-β-D-Glucuronide. Clin Transl Sci 2017; 10:271-279. [PMID: 28371445 PMCID: PMC5504481 DOI: 10.1111/cts.12458] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/27/2017] [Indexed: 01/05/2023] Open
Abstract
The oral multikinase inhibitor sorafenib undergoes extensive UGT1A9-mediated formation of sorafenib-β-D-glucuronide (SG). Using transporter-deficient mouse models, it was previously established that SG can be extruded into bile by ABCC2 or follow a liver-to-blood shuttling loop via ABCC3-mediated efflux into the systemic circulation, and subsequent uptake in neighboring hepatocytes by OATP1B-type transporters. Here we evaluated the possibility that this unusual process, called hepatocyte hopping, is also operational in humans and can be modulated through pharmacological inhibition. We found that SG transport by OATP1B1 or murine Oatp1b2 was effectively inhibited by rifampin, and that this agent can significantly increase plasma levels of SG in wildtype mice, but not in Oatp1b2-deficient animals. In human subjects receiving sorafenib, rifampin acutely increased the systemic exposure to SG. Our study emphasizes the need to consider hepatic handling of xenobiotic glucuronides in the design of drug-drug interaction studies of agents that undergo extensive phase II conjugation.
Collapse
Affiliation(s)
- S Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, Wytemaweg, Rotterdam, The Netherlands
| | - L van Doorn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Wytemaweg, Rotterdam, The Netherlands
| | - M A Phelps
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, Ohio State University, Columbus, Ohio, USA
| | - A A Gibson
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, Ohio State University, Columbus, Ohio, USA
| | - S Hu
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, Ohio State University, Columbus, Ohio, USA
| | - L Li
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - A Vasilyeva
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - G Du
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - P Hamberg
- Department of Internal Medicine, St. Franciscus Gasthuis, Rotterdam, The Netherlands
| | - Falm Eskens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Wytemaweg, Rotterdam, The Netherlands
| | - P de Bruijn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Wytemaweg, Rotterdam, The Netherlands
| | - A Sparreboom
- Department of Medical Oncology, Erasmus MC Cancer Institute, Wytemaweg, Rotterdam, The Netherlands.,Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, Ohio State University, Columbus, Ohio, USA
| | - Rhj Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Wytemaweg, Rotterdam, The Netherlands
| | - S D Baker
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
61
|
Preincubation-dependent and long-lasting inhibition of organic anion transporting polypeptide (OATP) and its impact on drug-drug interactions. Pharmacol Ther 2017; 177:67-80. [PMID: 28249706 DOI: 10.1016/j.pharmthera.2017.02.042] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Preincubation with cyclosporin A (CsA), a potent inhibitor of organic anion transporting polypeptide 1B1 (OATP1B1) and OATP1B3, enhanced its inhibitory effects on these transporters in vitro. A similar effect was observed upon preincubation with some other inhibitors. Removing these from the incubation media did not readily reverse the inhibition on OATP1B1 and OATP1B3. This preincubation-dependent long-lasting inhibition appeared to be related to CsA concentration in the cells in addition to that in the incubation media. Thus, we hypothesized that CsA inhibits OATP1B1 and OATP1B3 from inside (trans-inhibition) as well as outside (cis-inhibition) the cells and constructed the cis- and trans-inhibition model. The enhanced inhibitory effect of CsA on OATP1B1 observed after preincubation was quantitatively described using Ki,out and Ki,in as inhibition constants for cis- and trans-inhibitions, respectively. In addition, a long-lasting inhibition was also described by this model. Additional factors taken into consideration when simulating in vivo pharmacokinetic alterations by CsA are potential inhibition by AM1, a major metabolite of CsA, which has been reported to inhibit OATP1B1 and OATP1B3. Based on the physiologically based pharmacokinetic model incorporating trans- and cis-inhibition of OATP1B1 by CsA, the simulation showed that OATP1B1-mediated drug-drug interaction with CsA was suggested to be time-dependent also in vivo although further clinical studies are required for confirmation.
Collapse
|
62
|
Bi YA, Scialis RJ, Lazzaro S, Mathialagan S, Kimoto E, Keefer J, Zhang H, Vildhede AM, Costales C, Rodrigues AD, Tremaine LM, Varma MVS. Reliable Rate Measurements for Active and Passive Hepatic Uptake Using Plated Human Hepatocytes. AAPS JOURNAL 2017; 19:787-796. [DOI: 10.1208/s12248-017-0051-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/30/2017] [Indexed: 12/16/2022]
|
63
|
Shen H, Lai Y, Rodrigues AD. Organic Anion Transporter 2: An Enigmatic Human Solute Carrier. Drug Metab Dispos 2017; 45:228-236. [PMID: 27872146 DOI: 10.1124/dmd.116.072264] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/17/2016] [Indexed: 12/28/2022] Open
Abstract
As a member of the solute carrier 22A (SLC22A) family, organic anion transporter 2 (OAT2; SLC22A7) is emerging as an important drug transporter because of its expression in both the liver and kidney, two major eliminating organs, and its ability to transport not only a wide variety of xenobiotics but also numerous physiologically important endogenous compounds, like creatinine and cGMP. However, OAT2 has received relatively little attention compared with other OATs and solute carriers (SLCs), like organic cation transporters, sodium-dependent taurocholate cotransporting polypeptide, multidrug and toxin extrusion proteins, and organic anion-transporting polypeptides. Overall, the literature describing OAT2 is rapidly evolving, with numerous publications contradicting each other regarding the transport mechanism, tissue distribution, and transport of creatinine and cGMP, two important endogenous OAT2 substrates. Despite its status as a liver and kidney SLC, tools for assessing its activity and inhibition are lacking, and its role in drug disposition and elimination remains to be defined. The current review focuses on the available and emerging literature describing OAT2. We envision that OAT2 will gain more prominence as its expression, substrate, and inhibitor profile is investigated further and compared with other SLCs.
Collapse
Affiliation(s)
- Hong Shen
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| | - Yurong Lai
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| | - A David Rodrigues
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| |
Collapse
|
64
|
Shen H, Li W, Humphreys WG, Lai Y. Tenofovir Disoproxil Fumarate Is Not an Inhibitor of Human Organic Cation Transporter 1. J Pharmacol Exp Ther 2017; 360:341-342. [PMID: 28104831 DOI: 10.1124/jpet.116.238337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Hong Shen
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Wenying Li
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - W Griffith Humphreys
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Yurong Lai
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| |
Collapse
|
65
|
Physiologically Based Pharmacokinetic (PBPK) Modeling of Pitavastatin and Atorvastatin to Predict Drug-Drug Interactions (DDIs). Eur J Drug Metab Pharmacokinet 2016; 42:689-705. [DOI: 10.1007/s13318-016-0383-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
66
|
Kovacsics D, Patik I, Özvegy-Laczka C. The role of organic anion transporting polypeptides in drug absorption, distribution, excretion and drug-drug interactions. Expert Opin Drug Metab Toxicol 2016; 13:409-424. [PMID: 27783531 DOI: 10.1080/17425255.2017.1253679] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION The in vivo fate and effectiveness of a drug depends highly on its absorption, distribution, metabolism, excretion and toxicity (ADME-Tox). Organic anion transporting polypeptides (OATPs) are membrane proteins involved in the cellular uptake of various organic compounds, including clinically used drugs. Since OATPs are significant players in drug absorption and distribution, modulation of OATP function via pharmacotherapy with OATP substrates/inhibitors, or modulation of their expression, affects drug pharmacokinetics. Given their cancer-specific expression, OATPs may also be considered anticancer drug targets. Areas covered: We describe the human OATP family, discussing clinically relevant consequences of altered OATP function. We offer a critical analysis of published data on the role of OATPs in ADME and in drug-drug interactions, especially focusing on OATP1A2, 1B1, 1B3 and 2B1. Expert opinion: Four members of the OATP family, 1A2, 1B1, 1B3 and 2B1, have been characterized in detail. As biochemical and pharmacological knowledge on the other OATPs is lacking, it seems timely to direct research efforts towards developing the experimental framework needed to investigate the transport mechanism and substrate specificity of the poorly described OATPs. In addition, elucidating the role of OATPs in tumor development and therapy response are critical avenues for further research.
Collapse
Affiliation(s)
- Daniella Kovacsics
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Izabel Patik
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| | - Csilla Özvegy-Laczka
- a Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Budapest , Hungary
| |
Collapse
|
67
|
Abstract
Cells need to strictly control their internal milieu, a function which is performed by the plasma membrane. Selective passage of molecules across the plasma membrane is controlled by transport proteins. As the liver is the central organ for drug metabolism, hepatocytes are equipped with numerous drug transporters expressed at the plasma membrane. Drug disposition includes absorption, distribution, metabolism, and elimination of a drug and hence multiple passages of drugs and their metabolites across membranes. Consequently, understanding the exact mechanisms of drug transporters is essential both in drug development and in drug therapy. While many drug transporters are expressed in hepatocytes, and some of them are well characterized, several transporters have only recently been identified as new drug transporters. Novel powerful tools to deorphanize (drug) transporters are being applied and show promising results. Although a large set of tools are available for studying transport in vitro and in isolated cells, tools for studying transport in living organisms, including humans, are evolving now and rely predominantly on imaging techniques, e.g. positron emission tomography. Imaging is an area which, certainly in the near future, will provide important insights into "transporters at work" in vivo.
Collapse
Affiliation(s)
- Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, 8091, Switzerland
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| |
Collapse
|
68
|
Lai Y, Mandlekar S, Shen H, Holenarsipur VK, Langish R, Rajanna P, Murugesan S, Gaud N, Selvam S, Date O, Cheng Y, Shipkova P, Dai J, Humphreys WG, Marathe P. Coproporphyrins in Plasma and Urine Can Be Appropriate Clinical Biomarkers to Recapitulate Drug-Drug Interactions Mediated by Organic Anion Transporting Polypeptide Inhibition. J Pharmacol Exp Ther 2016; 358:397-404. [PMID: 27317801 DOI: 10.1124/jpet.116.234914] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/16/2016] [Indexed: 03/08/2025] Open
Abstract
In the present study, an open-label, three-treatment, three-period clinical study of rosuvastatin (RSV) and rifampicin (RIF) when administered alone and in combination was conducted in 12 male healthy subjects to determine if coproporphyrin I (CP-I) and coproporphyrin III (CP-III) could serve as clinical biomarkers for organic anion transporting polypeptide 1B1 (OATP1B1) and 1B3 that belong to the solute carrier organic anion gene subfamily. Genotyping of the human OATP1B1 gene was performed in all 12 subjects and confirmed absence of OATP1B1*5 and OATP1B1*15 mutations. Average plasma concentrations of CP-I and CP-III prior to drug administration were 0.91 ± 0.21 and 0.15 ± 0.04 nM, respectively, with minimum fluctuation over the three periods. CP-I was passively eliminated, whereas CP-III was actively secreted from urine. Administration of RSV caused no significant changes in the plasma and urinary profiles of CP-I and CP-III. RIF markedly increased the maximum plasma concentration (Cmax) of CP-I and CP-III by 5.7- and 5.4-fold (RIF) or 5.7- and 6.5-fold (RIF+RSV), respectively, as compared with the predose values. The area under the plasma concentration curves from time 0 to 24 h (AUC0-24h) of CP-I and CP-III with RIF and RSV increased by 4.0- and 3.3-fold, respectively, when compared with RSV alone. In agreement with this finding, Cmax and AUC0-24h of RSV increased by 13.2- and 5.0-fold, respectively, when RIF was coadministered. Collectively, we conclude that CP-I and CP-III in plasma and urine can be appropriate endogenous biomarkers specifically and reliably reflecting OATP inhibition, and thus the measurement of these molecules can serve as a useful tool to assess OATP drug-drug interaction liabilities in early clinical studies.
Collapse
Affiliation(s)
- Yurong Lai
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Sandhya Mandlekar
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Hong Shen
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Vinay K Holenarsipur
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Robert Langish
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Prabhakar Rajanna
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Senthilkumar Murugesan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Nilesh Gaud
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Sabariya Selvam
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Onkar Date
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Yaofeng Cheng
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Petia Shipkova
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Jun Dai
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - William G Humphreys
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| | - Punit Marathe
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey (Y.L., H.S., R.L., Y.C., P.S., J.D., W.G.H., P.M.); Bristol-Myers Squibb India Pvt. Ltd., Biocon Bristol-Myers Squibb Research and Development Center, Bangalore, India (Sa.M.); and Biocon BMS R&D Center, Syngene International Ltd., Bangalore, India (V.K.H., P.R., Se.M., N.G., S.S., O.D.)
| |
Collapse
|
69
|
Lechner C, Ishiguro N, Fukuhara A, Shimizu H, Ohtsu N, Takatani M, Nishiyama K, Washio I, Yamamura N, Kusuhara H. Impact of Experimental Conditions on the Evaluation of Interactions between Multidrug and Toxin Extrusion Proteins and Candidate Drugs. Drug Metab Dispos 2016; 44:1381-9. [PMID: 27271370 DOI: 10.1124/dmd.115.068163] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 06/03/2016] [Indexed: 11/22/2022] Open
Abstract
Multidrug and toxin extrusion transporters (MATEs) have a determining influence on the pharmacokinetic profiles of many drugs and are involved in several clinical drug-drug interactions (DDIs). Cellular uptake assays with recombinant cells expressing human MATE1 or MATE2-K are widely used to investigate MATE-mediated transport for DDI assessment; however, the experimental conditions and used test substrates vary among laboratories. We therefore initially examined the impact of three assay conditions that have been applied for MATE substrate and inhibitor profiling in the literature. One of the tested conditions resulted in significantly higher uptake rates of the three test substrates, [(14)C]metformin, [(3)H]thiamine, and [(3)H]1-methyl-4-phenylpyridinium (MPP(+)), but IC50 values of four tested MATE inhibitors varied only slightly among the three conditions (<2.5-fold difference). Subsequently, we investigated the uptake characteristics of the five MATE substrates: [(14)C]metformin, [(3)H]thiamine, [(3)H]MPP(+), [(3)H]estrone-3-sulfate (E3S), and rhodamine 123, as well as the impact of the used test substrate on the inhibition profiles of 10 MATE inhibitors at one selected assay condition. [(3)H]E3S showed atypical uptake characteristics compared with those observed with the other four substrates. IC50 values of the tested inhibitors were in a similar range (<4-fold difference) when [(14)C]metformin, [(3)H]thiamine, [(3)H]MPP(+), or [(3)H]E3S were used as substrates but were considerably higher with rhodamine 123 (9.8-fold and 4.1-fold differences compared with [(14)C]metformin with MATE1 and MATE2-K, respectively). This study demonstrated for the first time that the impact of assay conditions on IC50 determination is negligible, that kinetic characteristics differ among used test substrates, and that substrate-dependent inhibition exists for MATE1 and MATE2-K, giving valuable insight into the assessment of clinically relevant MATE-mediated DDIs in vitro.
Collapse
Affiliation(s)
- Christian Lechner
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Ayano Fukuhara
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Hidetada Shimizu
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Naoko Ohtsu
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Masahito Takatani
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Kotaro Nishiyama
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Ikumi Washio
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Norio Yamamura
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Hiroyuki Kusuhara
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan (C.L., N.I., A.F., H.S., N.O., M.T., K.N., I.W., N.Y.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| |
Collapse
|
70
|
Abstract. Drug Metab Rev 2016. [DOI: 10.1080/03602532.2016.1191843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
71
|
Shen H, Dai J, Liu T, Cheng Y, Chen W, Freeden C, Zhang Y, Humphreys WG, Marathe P, Lai Y. Coproporphyrins I and III as Functional Markers of OATP1B Activity: In Vitro and In Vivo Evaluation in Preclinical Species. J Pharmacol Exp Ther 2016; 357:382-93. [PMID: 26907622 DOI: 10.1124/jpet.116.232066] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/12/2016] [Indexed: 03/08/2025] Open
Abstract
Inhibition of organic anion-transporting polypeptide (OATP)1B function can lead to serious clinical drug-drug interactions, thus a thorough evaluation of the potential for this type of interaction must be completed during drug development. Therefore, sensitive and specific biomarkers for OATP function that could be used in conjunction with clinical studies are currently in demand. In the present study, preclinical evaluations were conducted to characterize the suitability of coproporphyrins (CPs) I and III as markers of hepatic OATP functional activity. Active uptake of CPs I and III was observed in human embryonic kidney (HEK) 293 cells singly expressing human OATP1B1 (hOATP1B1), hOATP1B3, cynomolgus monkey OATP1B1 (cOATP1B1), or cOATP1B3, as well as human and monkey hepatocytes. Cyclosporin A (100 mg/kg, oral) markedly increased the area under the curve (AUC) plasma concentrations of CPs I and III by 2.6- and 5.2-fold, while rifampicin (15 mg/kg, oral) increased the AUCs by 2.7- and 3.6-fold, respectively. As the systemic exposure increased, the excretion of both isomers in urine rose from 1.6- to 4.3-fold in monkeys. In agreement with this finding, the AUC of rosuvastatin (RSV) in cynomolgus monkeys increased when OATP1B inhibitors were coadministered. In Oatp1a/1b gene cluster knockout mice (Oatp1a/1b(-/-)), CPs in plasma and urine were significantly increased compared with wild-type animals (7.1- to 18.4-fold; P < 0.001), which were also in agreement with the changes in plasma RSV exposure (14.6-fold increase). We conclude that CPs I and III in plasma and urine are novel endogenous biomarkers reflecting hepatic OATP function, and the measurements have the potential to be incorporated into the design of early clinical evaluation.
Collapse
Affiliation(s)
- Hong Shen
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Jun Dai
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Tongtong Liu
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Yaofeng Cheng
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Weiqi Chen
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Chris Freeden
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Yingru Zhang
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - W Griffith Humphreys
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Punit Marathe
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Yurong Lai
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, New Jersey
| |
Collapse
|
72
|
Vermeer LMM, Isringhausen CD, Ogilvie BW, Buckley DB. Evaluation of Ketoconazole and Its Alternative Clinical CYP3A4/5 Inhibitors as Inhibitors of Drug Transporters: The In Vitro Effects of Ketoconazole, Ritonavir, Clarithromycin, and Itraconazole on 13 Clinically-Relevant Drug Transporters. Drug Metab Dispos 2016; 44:453-9. [PMID: 26668209 DOI: 10.1124/dmd.115.067744] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/11/2015] [Indexed: 01/18/2023] Open
Abstract
Ketoconazole is a potent CYP3A4/5 inhibitor and, until recently, recommended by the Food and Drug Administration (FDA) and the European Medicines Agency as a strong CYP3A4/5 inhibitor in clinical drug-drug interaction (DDI) studies. Ketoconazole sporadically causes liver injury or adrenal insufficiency. Because of this, the FDA and European Medicines Agency recommended suspension of ketoconazole use in DDI studies in 2013. The FDA specifically recommended use of clarithromycin or itraconazole as alternative strong CYP3A4/5 inhibitors in clinical DDI studies, but many investigators have also used ritonavir as an alternative. Although the effects of these clinical CYP3A4/5 inhibitors on other CYPs are largely established, reports on the effects on the broad range of drug transporter activities are sparse. In this study, the inhibitory effects of ketoconazole, clarithromycin, ritonavir, and itraconazole (and its CYP3A4-inhibitory metabolites, hydroxy-, keto-, and N-desalkyl itraconazole) toward 13 drug transporters (OATP1B1, OATP1B3, OAT1, OAT3, OCT1, OCT2, MATE1, MATE2-K, P-gp, BCRP, MRP2, MRP3, and BSEP) were systematically assessed in transporter-expressing HEK-293 cell lines or membrane vesicles. In vitro findings were translated into clinical context with the basic static model approaches outlined by the FDA in its 2012 draft guidance on DDIs. The results indicate that, like ketoconazole, the alternative clinical CYP3A4/5 inhibitors ritonavir, clarithromycin, and itraconazole each have unique transporter inhibition profiles. None of the alternatives to ketoconazole provided a clean inhibition profile toward the 13 drug transporters evaluated. The results provide guidance for the selection of clinical CYP3A4/5 inhibitors when transporters are potentially involved in a victim drug's pharmacokinetics.
Collapse
|
73
|
Riley RJ, Foley SA, Barton P, Soars MG, Williamson B. Hepatic drug transporters: the journey so far. Expert Opin Drug Metab Toxicol 2016; 12:201-16. [PMID: 26670591 DOI: 10.1517/17425255.2016.1132308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The key role of transporter biology in both the manifestation and treatment of disease is now firmly established. Experiences of sub-optimal drug exposure due to drug-transporter interplay have supported incorporation of studies aimed at understanding the interactions between compounds and drug transporters much earlier in drug discovery. While drug transporters can impact the most pivotal pharmacokinetic parameter with respect to human dose and exposure projections, clearance, at a renal or hepatobiliary level, the latter will form the focus of this perspective. AREAS COVERED A synopsis of guidelines on which transporters to study together with an overview of the currently available toolkit is presented. A perspective on when to conduct studies with various hepatic transporters is also provided together with structural "alerts" which should prompt early investigation. EXPERT OPINION Great progress has been made in individual laboratories and via consortia to understand the role of drug transporters in disease, drug disposition, drug-drug interactions and toxicity. A systematic analysis of the value posed by the available approaches and an inter-lab comparison now seems warranted. The emerging ability to use physico-chemical properties to guide future screening cascades promises to revolutionise the efficiency of early drug discovery.
Collapse
Affiliation(s)
| | | | - P Barton
- b School of Life Sciences , University of Nottingham , Nottingham , UK
| | - M G Soars
- c Drug Metabolism and Pharmacokinetics , Bristol-Myers Squibb , Wallingford , CT , USA
| | | |
Collapse
|
74
|
Izumi S, Nozaki Y, Komori T, Takenaka O, Maeda K, Kusuhara H, Sugiyama Y. Investigation of Fluorescein Derivatives as Substrates of Organic Anion Transporting Polypeptide (OATP) 1B1 To Develop Sensitive Fluorescence-Based OATP1B1 Inhibition Assays. Mol Pharm 2016; 13:438-48. [DOI: 10.1021/acs.molpharmaceut.5b00664] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Saki Izumi
- Drug Metabolism
and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co.
Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yoshitane Nozaki
- Drug Metabolism
and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co.
Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Takafumi Komori
- Drug Metabolism
and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co.
Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Osamu Takenaka
- Modeling & Simulation, Clinical Pharmacology, Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo 112-8088, Japan
| | - Kazuya Maeda
- Laboratory
of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroyuki Kusuhara
- Laboratory
of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuichi Sugiyama
- Sugiyama
Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama-shi, Kanagawa 230-0045, Japan
| |
Collapse
|
75
|
Lin L, Yee SW, Kim RB, Giacomini KM. SLC transporters as therapeutic targets: emerging opportunities. Nat Rev Drug Discov 2015; 14:543-60. [PMID: 26111766 DOI: 10.1038/nrd4626] [Citation(s) in RCA: 563] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Solute carrier (SLC) transporters - a family of more than 300 membrane-bound proteins that facilitate the transport of a wide array of substrates across biological membranes - have important roles in physiological processes ranging from the cellular uptake of nutrients to the absorption of drugs and other xenobiotics. Several classes of marketed drugs target well-known SLC transporters, such as neurotransmitter transporters, and human genetic studies have provided powerful insight into the roles of more-recently characterized SLC transporters in both rare and common diseases, indicating a wealth of new therapeutic opportunities. This Review summarizes knowledge on the roles of SLC transporters in human disease, describes strategies to target such transporters, and highlights current and investigational drugs that modulate SLC transporters, as well as promising drug targets.
Collapse
Affiliation(s)
- Lawrence Lin
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94158, USA
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94158, USA
| | - Richard B Kim
- Division of Clinical Pharmacology, Department of Medicine, University of Western Ontario, London Health Science Centre, London, Ontario N6A 5A5, Canada
| | - Kathleen M Giacomini
- 1] Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94158, USA. [2] Institute for Human Genetics, University of California San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
76
|
Chu X, Shih SJ, Shaw R, Hentze H, Chan GH, Owens K, Wang S, Cai X, Newton D, Castro-Perez J, Salituro G, Palamanda J, Fernandis A, Ng CK, Liaw A, Savage MJ, Evers R. Evaluation of cynomolgus monkeys for the identification of endogenous biomarkers for hepatic transporter inhibition and as a translatable model to predict pharmacokinetic interactions with statins in humans. Drug Metab Dispos 2015; 43:851-63. [PMID: 25813937 DOI: 10.1124/dmd.115.063347] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/26/2015] [Indexed: 12/31/2022] Open
Abstract
Inhibition of hepatic transporters such as organic anion transporting polypeptides (OATPs) 1B can cause drug-drug interactions (DDIs). Determining the impact of perpetrator drugs on the plasma exposure of endogenous substrates for OATP1B could be valuable to assess the risk for DDIs early in drug development. As OATP1B orthologs are well conserved between human and monkey, we assessed in cynomolgus monkeys the endogenous OATP1B substrates that are potentially suitable to assess DDI risk in humans. The effect of rifampin (RIF), a potent inhibitor for OATP1B, on plasma exposure of endogenous substrates of hepatic transporters was measured. From the 18 biomarkers tested, RIF (18 mg/kg, oral) caused significant elevation of plasma unconjugated and conjugated bilirubin, which may be attributed to inhibition of cOATP1B1 and cOATP1B3 based on in vitro to in vivo extrapolation analysis. To further evaluate whether cynomolgus monkeys are a suitable translational model to study OATP1B-mediated DDIs, we determined the inhibitory effect of RIF on in vitro transport and pharmacokinetics of rosuvastatin (RSV) and atorvastatin (ATV). RIF strongly inhibited the uptake of RSV and ATV by cOATP1B1 and cOATP1B3 in vitro. In agreement with clinical observations, RIF (18 mg/kg, oral) significantly decreased plasma clearance and increased the area under the plasma concentration curve (AUC) of intravenously administered RSV by 2.8- and 2.7-fold, and increased the AUC and maximum plasma concentration of orally administered RSV by 6- and 10.3-fold, respectively. In contrast to clinical findings, RIF did not significantly increase plasma exposure of either intravenous or orally administered ATV, indicating species differences in the rate-limiting elimination pathways.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Shian-Jiun Shih
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Rachel Shaw
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Hannes Hentze
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Grace H Chan
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Karen Owens
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Shubing Wang
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Xiaoxin Cai
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Deborah Newton
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Jose Castro-Perez
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Gino Salituro
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Jairam Palamanda
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Aaron Fernandis
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Choon Keow Ng
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Andy Liaw
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Mary J Savage
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Raymond Evers
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| |
Collapse
|
77
|
Prueksaritanont T, Chu X, Evers R, Klopfer SO, Caro L, Kothare PA, Dempsey C, Rasmussen S, Houle R, Chan G, Cai X, Valesky R, Fraser IP, Stoch SA. Pitavastatin is a more sensitive and selective organic anion-transporting polypeptide 1B clinical probe than rosuvastatin. Br J Clin Pharmacol 2015; 78:587-98. [PMID: 24617605 DOI: 10.1111/bcp.12377] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/05/2014] [Indexed: 12/14/2022] Open
Abstract
AIMS Rosuvastatin and pitavastatin have been proposed as probe substrates for the organic anion-transporting polypeptide (OATP) 1B, but clinical data on their relative sensitivity and selectivity to OATP1B inhibitors are lacking. A clinical study was therefore conducted to determine their relative suitability as OATP1B probes using single oral (PO) and intravenous (IV) doses of the OATP1B inhibitor rifampicin, accompanied by a comprehensive in vitro assessment of rifampicin inhibitory potential on statin transporters. METHODS The clinical study comprised of two separate panels of eight healthy subjects. In each panel, subjects were randomized to receive a single oral dose of rosuvastatin (5 mg) or pitavastatin (1 mg) administered alone, concomitantly with rifampicin (600 mg) PO or IV. The in vitro transporter studies were performed using hepatocytes and recombinant expression systems. RESULTS Rifampicin markedly increased exposures of both statins, with greater differential increases after PO vs. IV rifampicin only for rosuvastatin. The magnitudes of the increases in area under the plasma concentration-time curve were 5.7- and 7.6-fold for pitavastatin and 4.4- and 3.3-fold for rosuvastatin, after PO and IV rifampicin, respectively. In vitro studies showed that rifampicin was an inhibitor of OATP1B1 and OATP1B3, breast cancer resistance protein and multidrug resistance protein 2, but not of organic anion transporter 3. CONCLUSIONS The results indicate that pitavastatin is a more sensitive and selective and thus preferred clinical OATP1B probe substrate than rosuvastatin, and that a single IV dose of rifampicin is a more selective OATP1B inhibitor than a PO dose.
Collapse
|
78
|
Le Vee M, Jouan E, Stieger B, Lecureur V, Fardel O. Regulation of human hepatic drug transporter activity and expression by diesel exhaust particle extract. PLoS One 2015; 10:e0121232. [PMID: 25803276 PMCID: PMC4372591 DOI: 10.1371/journal.pone.0121232] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/28/2015] [Indexed: 12/31/2022] Open
Abstract
Diesel exhaust particles (DEPs) are common environmental air pollutants primarily affecting the lung. DEPs or chemicals adsorbed on DEPs also exert extra-pulmonary effects, including alteration of hepatic drug detoxifying enzyme expression. The present study was designed to determine whether organic DEP extract (DEPe) may target hepatic drug transporters that contribute in a major way to drug detoxification. Using primary human hepatocytes and transporter-overexpressing cells, DEPe was first shown to strongly inhibit activities of the sinusoidal solute carrier (SLC) uptake transporters organic anion-transporting polypeptides (OATP) 1B1, 1B3 and 2B1 and of the canalicular ATP-binding cassette (ABC) efflux pump multidrug resistance-associated protein 2, with IC50 values ranging from approximately 1 to 20 μg/mL and relevant to environmental exposure situations. By contrast, 25 μg/mL DEPe failed to alter activities of the SLC transporter organic cation transporter (OCT) 1 and of the ABC efflux pumps P-glycoprotein and bile salt export pump (BSEP), whereas it only moderately inhibited those of sodium taurocholate co-transporting polypeptide and of breast cancer resistance protein (BCRP). Treatment by 25 μg/mL DEPe was next demonstrated to induce expression of BCRP at both mRNA and protein level in cultured human hepatic cells, whereas it concomitantly repressed mRNA expression of various transporters, including OATP1B3, OATP2B1, OCT1 and BSEP. Such changes in transporter expression were found to be highly correlated to those caused by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a reference activator of the aryl hydrocarbon receptor (AhR) pathway. This suggests that DEPe, which is enriched in known ligands of AhR like polycyclic aromatic hydrocarbons, alters drug transporter expression via activation of the AhR cascade. Taken together, these data established human hepatic transporters as targets of organic chemicals containing in DEPs, which may contribute to their systemic effects through impairing hepatic transport of endogenous compound or drug substrates of these transporters.
Collapse
Affiliation(s)
- Marc Le Vee
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Elodie Jouan
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Valérie Lecureur
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Olivier Fardel
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France
- Pôle Biologie, Centre Hospitalier Universitaire, 2 rue Henri Le Guilloux, 35033 Rennes, France
- * E-mail:
| |
Collapse
|
79
|
Izumi S, Nozaki Y, Maeda K, Komori T, Takenaka O, Kusuhara H, Sugiyama Y. Investigation of the impact of substrate selection on in vitro organic anion transporting polypeptide 1B1 inhibition profiles for the prediction of drug-drug interactions. Drug Metab Dispos 2015; 43:235-47. [PMID: 25414411 DOI: 10.1124/dmd.114.059105] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
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-17β-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.
Collapse
Affiliation(s)
- Saki Izumi
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co. Ltd., Ibaraki, Japan (S.I., Y.N., T.K.); Pharmacokinetics and Pharmacodynamics, Morphotek Inc., Exton, Pennsylvania (O.T.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (Y.S.)
| | - Yoshitane Nozaki
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co. Ltd., Ibaraki, Japan (S.I., Y.N., T.K.); Pharmacokinetics and Pharmacodynamics, Morphotek Inc., Exton, Pennsylvania (O.T.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (Y.S.)
| | - Kazuya Maeda
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co. Ltd., Ibaraki, Japan (S.I., Y.N., T.K.); Pharmacokinetics and Pharmacodynamics, Morphotek Inc., Exton, Pennsylvania (O.T.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (Y.S.)
| | - Takafumi Komori
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co. Ltd., Ibaraki, Japan (S.I., Y.N., T.K.); Pharmacokinetics and Pharmacodynamics, Morphotek Inc., Exton, Pennsylvania (O.T.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (Y.S.)
| | - Osamu Takenaka
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co. Ltd., Ibaraki, Japan (S.I., Y.N., T.K.); Pharmacokinetics and Pharmacodynamics, Morphotek Inc., Exton, Pennsylvania (O.T.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (Y.S.)
| | - Hiroyuki Kusuhara
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co. Ltd., Ibaraki, Japan (S.I., Y.N., T.K.); Pharmacokinetics and Pharmacodynamics, Morphotek Inc., Exton, Pennsylvania (O.T.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (Y.S.)
| | - Yuichi Sugiyama
- Drug Metabolism and Pharmacokinetics Japan, Tsukuba Research Laboratories, Eisai Co. Ltd., Ibaraki, Japan (S.I., Y.N., T.K.); Pharmacokinetics and Pharmacodynamics, Morphotek Inc., Exton, Pennsylvania (O.T.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (Y.S.)
| |
Collapse
|
80
|
Marada VVVR, Flörl S, Kühne A, Burckhardt G, Hagos Y. Interaction of human organic anion transporter polypeptides 1B1 and 1B3 with antineoplastic compounds. Eur J Med Chem 2015; 92:723-31. [PMID: 25618019 DOI: 10.1016/j.ejmech.2015.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/08/2014] [Accepted: 01/07/2015] [Indexed: 12/22/2022]
Abstract
Antineoplastic compounds are used in the treatment of a variety of cancers. The effectiveness of an antineoplastic compound to exert its activity is largely dependent on transport proteins involved in the entry of the compound into the cells, and those which drive it out of the cell. Organic anion transporting polypeptide 1B1 (OATP1B1) and organic anion transporting polypeptide 1B3 (OATP1B3), belonging to the SLCO family of proteins, are specifically expressed in the sinusoidal membranes of the liver, and are known to interact with a variety of drugs. The present study deals with the interaction of these proteins with antineoplastic compounds routinely used in cancer chemotherapy. The proteins OATP1B1 and OATP1B3 were functionally characterized in stably transfected human embryonic kidney cells using [(3)H] labeled estrone 3-sulfate and [(3)H] labeled cholecystokinin octapeptide (CCK-8) as substrates, respectively. Substrate uptake experiments performed in the presence of antineoplastic compounds showed that vinblastine and paclitaxel strongly interacted with the OATP1B1 with Ki values of 10.2 μM and 0.84 μM, respectively. OATP1B3 showed highly significant interactions with a variety of antineoplastic compounds including chlorambucil, mitoxantrone, vinblastine, vincristine, paclitaxel and etoposide, with Ki values of 40.6 μM, 3.2 μM, 15.9 μM, 30.6 μM, 1.8 μM and 13.5 μM, respectively. We report several novel interactions of the transporter proteins OATP1B1 and OATP1B3 highlighting the need to investigate their role in drug-drug interactions and cancer chemotherapy.
Collapse
Affiliation(s)
- Venkata V V R Marada
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Saskia Flörl
- PortaCellTec Biosciences GmbH, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Annett Kühne
- PortaCellTec Biosciences GmbH, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Gerhard Burckhardt
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany.
| | - Yohannes Hagos
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany; PortaCellTec Biosciences GmbH, Humboldtallee 23, 37073 Göttingen, Germany.
| |
Collapse
|
81
|
Maeda K. Organic Anion Transporting Polypeptide (OATP)1B1 and OATP1B3 as Important Regulators of the Pharmacokinetics of Substrate Drugs. Biol Pharm Bull 2015; 38:155-68. [DOI: 10.1248/bpb.b14-00767] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences,
The University of Tokyo
| |
Collapse
|
82
|
Higgins JW, Ke AB, Zamek-Gliszczynski MJ. Clinical CYP3A inhibitor alternatives to ketoconazole, clarithromycin and itraconazole, are not transported into the liver by hepatic organic anion transporting polypeptides and organic cation transporter 1. Drug Metab Dispos 2014; 42:1780-4. [PMID: 25106415 DOI: 10.1124/dmd.114.058784] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Ketoconazole is no longer available for clinical determination of worst-case victim drug-drug interaction (DDI) potential for cytochrome P450 3A (CYP3A)-substrate drugs; clarithromycin and itraconazole are the proposed replacements. Ketoconazole DDIs are described by unbound systemic exposures due to absence of carrier-facilitated hepatic uptake, but this aspect of clarithromycin and itraconazole disposition has not been investigated. At present, transport of clarithromycin, itraconazole, and hydroxyitraconazole by hepatic organic anion transporting polypeptides (OATPs) and organic cation transporter 1 (OCT1) was examined in vitro and in vivo. As for ketoconazole, uptake of clarithromycin, itraconazole, and hydroxyitraconazole into OATP1B1, OATP1B3, OATP2B1, and OCT1 expressing human embryonic kidney 293 (HEK293) cells was not greater than in vector controls. Uptake into these HEK293 cells and human hepatocytes was not impaired by the prototypical OATP, OCT, and sodium/taurocholate cotransporting polypeptide inhibitors bromosulfophthalein, imipramine, and taurocholate, respectively. In contrast, uptake of the positive controls, atorvastatin for OATPs and metformin for OCT1, was significantly enhanced by relevant transporter expression, and uptake into both these HEK293 cells and human hepatocytes was significantly impaired by prototypical inhibitors. In Oatp1a/1b gene cluster knockout mice, which lack the major hepatic Oatps, and in Oct1/2 knockout mice, ketoconazole, clarithromycin, itraconazole, and hydroxyitraconazole oral exposure was not increased, and the liver-to-blood partition coefficient (Kp) was not decreased. By contrast relative to wild-type mice, in Oatp1a/1b- and Oct1/2-knockout mice, atorvastatin and metformin oral exposure was significantly increased, and liver Kp was significantly decreased. The present studies provide in vitro and in vivo evidence that, like ketoconazole, clarithromycin, itraconazole, and hydroxyitraconazole are not transported into the liver by hepatic uptake transporters, including OATPs and OCT1.
Collapse
Affiliation(s)
- J William Higgins
- Drug Disposition, Lilly Research Laboratories, Indianapolis, Indiana
| | - Alice B Ke
- Drug Disposition, Lilly Research Laboratories, Indianapolis, Indiana
| | | |
Collapse
|
83
|
Johnston RA, Rawling T, Chan T, Zhou F, Murray M. Selective inhibition of human solute carrier transporters by multikinase inhibitors. Drug Metab Dispos 2014; 42:1851-7. [PMID: 25165131 DOI: 10.1124/dmd.114.059097] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Solute carrier (SLC) transporters regulate the cellular influx and disposition of endogenous and xenobiotic compounds, including anticancer agents such as the multikinase inhibitors (MKIs). Recent evidence suggests that MKIs may also inhibit SLC-dependent transport of coadministered drugs, although present information on the relative susceptibilities of multiple SLC transporters is limited. This study evaluated 18 MKI drugs and metabolites as inhibitors of prototypic substrate uptake by 13 SLC transporters that were overexpressed in human embryonic kidney cells. Organic anion transporting polypeptides (OATPs) 1A2, 1B3, and 2B1, organic anion transporter 3 (OAT3), and organic cation transporter 1 (OCT1) were inhibited by most MKIs, whereas substrate uptake by OATP1B1, OAT1, 2, and 4, OCT2 and 3, and organic zwitterion/cation transporter 1 (OCTN1) was less susceptible to inhibition; OCTN2 was also inhibited by cediranib. In further studies, IC50 values were determined for the most effective MKIs, and erlotinib and cediranib were found to be potent competitive inhibitors of OATP2B1 (Ki = 41 nM) and OATP1A2 (Ki = 33 nM), respectively. From predictive approaches, several MKI-SLC interactions were found to be of potential in vivo significance.
Collapse
Affiliation(s)
- Rosie A Johnston
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, Sydney Medical School (R.A.J., M.M.) and Faculty of Pharmacy (T.C., F.Z.), University of Sydney, New South Wales, Australia; and Discipline of Pharmacy, Graduate School of Health, University of Technology, Ultimo, New South Wales, Australia (T.R.)
| | - Tristan Rawling
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, Sydney Medical School (R.A.J., M.M.) and Faculty of Pharmacy (T.C., F.Z.), University of Sydney, New South Wales, Australia; and Discipline of Pharmacy, Graduate School of Health, University of Technology, Ultimo, New South Wales, Australia (T.R.)
| | - Ting Chan
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, Sydney Medical School (R.A.J., M.M.) and Faculty of Pharmacy (T.C., F.Z.), University of Sydney, New South Wales, Australia; and Discipline of Pharmacy, Graduate School of Health, University of Technology, Ultimo, New South Wales, Australia (T.R.)
| | - Fanfan Zhou
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, Sydney Medical School (R.A.J., M.M.) and Faculty of Pharmacy (T.C., F.Z.), University of Sydney, New South Wales, Australia; and Discipline of Pharmacy, Graduate School of Health, University of Technology, Ultimo, New South Wales, Australia (T.R.)
| | - Michael Murray
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, Sydney Medical School (R.A.J., M.M.) and Faculty of Pharmacy (T.C., F.Z.), University of Sydney, New South Wales, Australia; and Discipline of Pharmacy, Graduate School of Health, University of Technology, Ultimo, New South Wales, Australia (T.R.)
| |
Collapse
|
84
|
Zhang Q, Zhang Y, Diamond S, Boer J, Harris JJ, Li Y, Rupar M, Behshad E, Gardiner C, Collier P, Liu P, Burn T, Wynn R, Hollis G, Yeleswaram S. The Janus kinase 2 inhibitor fedratinib inhibits thiamine uptake: a putative mechanism for the onset of Wernicke's encephalopathy. Drug Metab Dispos 2014; 42:1656-62. [PMID: 25063672 DOI: 10.1124/dmd.114.058883] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The clinical development of fedratinib, a Janus kinase (JAK2) inhibitor, was terminated after reports of Wernicke's encephalopathy in myelofibrosis patients. Since Wernicke's encephalopathy is induced by thiamine deficiency, investigations were conducted to probe possible mechanisms through which fedratinib may lead to a thiamine-deficient state. In vitro studies indicate that fedratinib potently inhibits the carrier-mediated uptake and transcellular flux of thiamine in Caco-2 cells, suggesting that oral absorption of dietary thiamine is significantly compromised by fedratinib dosing. Transport studies with recombinant human thiamine transporters identified the individual human thiamine transporter (hTHTR2) that is inhibited by fedratinib. Inhibition of thiamine uptake appears unique to fedratinib and is not shared by marketed JAK inhibitors, and this observation is consistent with the known structure-activity relationship for the binding of thiamine to its transporters. The results from these studies provide a molecular basis for the development of Wernicke's encephalopathy upon fedratinib treatment and highlight the need to evaluate interactions of investigational drugs with nutrient transporters in addition to classic xenobiotic transporters.
Collapse
Affiliation(s)
| | - Yan Zhang
- Incyte Corporation, Wilmington, Delaware
| | | | - Jason Boer
- Incyte Corporation, Wilmington, Delaware
| | | | - Yu Li
- Incyte Corporation, Wilmington, Delaware
| | - Mark Rupar
- Incyte Corporation, Wilmington, Delaware
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Li R, Barton HA, Varma MV. Prediction of Pharmacokinetics and Drug–Drug Interactions When Hepatic Transporters are Involved. Clin Pharmacokinet 2014; 53:659-78. [DOI: 10.1007/s40262-014-0156-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|