1
|
Gulnaz A, Lee KR, Kang MJ, Chang JE, Chae YJ. Roles of breast cancer resistance protein and organic anion transporting polypeptide 2B1 in gastrointestinal toxicity induced by SN-38 under inflammatory conditions. Toxicol Lett 2024; 394:57-65. [PMID: 38423481 DOI: 10.1016/j.toxlet.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Drug transporters are among the factors that determine the pharmacokinetic profiles after drug administration. In this study, we investigated the roles of drug transporters involved in transport of SN-38, which is an active metabolite of irinotecan, in the intestine under inflammatory conditions in vitro and determined their functional consequences. The expression alterations of breast cancer resistance protein (BCRP) and organic anion transporting polypeptide (OATP) 2B1 were determined at the mRNA and protein levels, and the subsequent functional alterations were evaluated via an accumulation study with the representative transporter substrates [prazosin and dibromofluorescein (DBF)] and SN-38. We also determined the cytotoxicity of SN-38 under inflammatory conditions. Decreased BCRP expression and increased OATP2B1 expression were observed under inflammatory conditions in vitro, which led to altered accumulation profiles of prazosin, DBF, and SN-38, and the subsequent cytotoxic profiles of SN-38. Treatment with rifampin or novobiocin supported the significant roles of BCRP and OATP2B1 in the transport and cytotoxic profile of SN-38. Collectively, these results suggest that BCRP and OATP2B1 are involved in the increased cytotoxicity of SN-38 under inflammatory conditions in vitro. Further comprehensive research is warranted to completely understand SN-38-induced gastrointestinal cytotoxicity and aid in the successful treatment of cancer with irinotecan.
Collapse
Affiliation(s)
- Aneela Gulnaz
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Bioscience, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Min-Ji Kang
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Ji-Eun Chang
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea.
| |
Collapse
|
2
|
Baze A, Wiss L, Horbal L, Biemel K, Asselin L, Richert L. Comparison of in vitro thyroxine (T4) metabolism between Wistar rat and human hepatocyte cultures. Toxicol In Vitro 2024; 96:105763. [PMID: 38142784 DOI: 10.1016/j.tiv.2023.105763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/24/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
In vitro assays remain relatively new in exploring human relevance of liver, in particular nuclear receptor-mediated perturbations of the hypothalamus-pituitary-thyroid axis seen in rodents, mainly in the rat. Consistent with in vivo data, we confirm that thyroid hormone thyroxine metabolism was 9 times higher in primary rat hepatocytes (PRH) than in primary human hepatocytes (PHH) cultured in a 2D sandwich (2Dsw) configuration. In addition, thyroxine glucuronide (T4-G) was by far the major metabolite formed in both species (99.1% in PRH and 69.7% in PHH) followed by thyroxine sulfate (T4-S, 0.7% in PRH and 18.1% in PHH) and triiodothyronine/reverse triiodothyronine (T3/rT3, 0.2% in PRH and 12.2% in PHH). After a 7-day daily exposure to orphan receptor-mediated liver inducers, T4 metabolism was strongly increased in PRH, almost exclusively through increased T4-G formation. These results were consistent with the inductions of glucuronosyltransferase Ugt2b1 and canalicular transporter Mrp2. PHH also responded to activation of the three nuclear receptors, with mainly induction of glucuronosyltransferase UGT1A1 and canalicular transporter MRP2. Despite this, T4 disappearance rate and secreted T4 metabolites were only slightly increased in PHH. Overall, our data highlight that cryopreserved hepatocytes in 2Dsw culture allowing long-term exposure and species comparison are of major interest in improving liver-mediated human safety assessment.
Collapse
Affiliation(s)
- Audrey Baze
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Lucille Wiss
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Liliia Horbal
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrüken, Germany
| | - Klaus Biemel
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrüken, Germany
| | - Laure Asselin
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Lysiane Richert
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France; Zylan SAS, 8 rue de la Haute Corniche, 67210 Obernai, France.
| |
Collapse
|
3
|
Robbins JA, Menzel K, Lassman M, Zhao T, Fancourt C, Chu X, Mostoller K, Witter R, Marceau West R, Stoch SA, McCrea JB, Iwamoto M. Acute and Chronic Effects of Rifampin on Letermovir Suggest Transporter Inhibition and Induction Contribute to Letermovir Pharmacokinetics. Clin Pharmacol Ther 2021; 111:664-675. [PMID: 34888851 DOI: 10.1002/cpt.2510] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 12/06/2021] [Indexed: 11/06/2022]
Abstract
Rifampin has acute inhibitory and chronic inductive effects that can cause complex drug-drug interactions. Rifampin inhibits transporters including organic-anion-transporting polypeptide (OATP)1B and P-glycoprotein (P-gp), and induces enzymes and transporters including cytochrome P450 3A, UDP-glucuronosyltransferase (UGT)1A, and P-gp. This study aimed at separating inhibitory and inductive effects of rifampin on letermovir disposition and elimination (indicated for cytomegalovirus prophylaxis in hematopoietic stem cell transplant recipients). Letermovir is a substrate of UGT1A1/3, P-gp, and OATP1B, with its clearance primarily mediated by OATP1B. Letermovir (single-dose) administered with rifampin (single-dose) resulted in increased letermovir exposure through transporter inhibition. Chronic coadministration with rifampin (inhibition plus potential OATP1B induction) resulted in modestly decreased letermovir exposure versus letermovir alone. Letermovir administered 24 hours after last rifampin dose (potential OATP1B induction) resulted in markedly decreased letermovir exposure. These data suggest rifampin may induce transporters that clear letermovir; the modestly reduced letermovir exposure with chronic rifampin coadministration likely reflects the net effect of inhibition and induction. OATP1B endogenous biomarkers coproporphyrin (CP) I and glycochenodeoxycholic acid-sulfate (GCDCA-S) were also analyzed; their exposures increased after single-dose rifampin plus letermovir, consistent with OATP1B inhibition and prior reports of inhibition by rifampin alone. CP I and GCDCA-S exposures were substantially reduced with letermovir administered 24 hours after the last dose of rifampin versus letermovir plus chronic rifampin coadministration, This study suggests that OATP1B induction may contribute to reduced letermovir exposure after chronic rifampin administration, although given the complexity of letermovir disposition, alternative mechanisms are not fully excluded.
Collapse
Affiliation(s)
| | | | | | - Tian Zhao
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Classification of drugs for evaluating drug interaction in drug development and clinical management. Drug Metab Pharmacokinet 2021; 41:100414. [PMID: 34666290 DOI: 10.1016/j.dmpk.2021.100414] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 12/22/2022]
Abstract
During new drug development, clinical drug interaction studies are carried out in accordance with the mechanism of potential drug interactions evaluated by in vitro studies. The obtained information should be provided efficiently to medical experts through package inserts and various information materials after the drug's launch. A recently updated Japanese guideline presents general procedures that are considered scientifically valid at the present moment. In this review, we aim to highlight the viewpoints of the Japanese guideline and enumerate drugs that were involved or are anticipated to be involved in evident pharmacokinetic drug interactions and classify them by their clearance pathway and potential intensity based on systematic reviews of the literature. The classification would be informative for designing clinical studies during the development stage, and the appropriate management of drug interactions in clinical practice.
Collapse
|
5
|
Fan X, Bai J, Hu M, Xu Y, Zhao S, Sun Y, Wang B, Hu J, Li Y. Drug interaction study of flavonoids toward OATP1B1 and their 3D structure activity relationship analysis for predicting hepatoprotective effects. Toxicology 2020; 437:152445. [PMID: 32259555 DOI: 10.1016/j.tox.2020.152445] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/14/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022]
Abstract
Organic anion transporting polypeptide 1B1 (OATP1B1), a liver-specific uptake transporter, was associated with drug induced liver injury (DILI). Screening and identifying potent OATP1B1 inhibitors with little toxicity is of great value in reducing OATP1B1-mediated DILI. Flavonoids are a group of polyphenols ubiquitously present in vegetables, fruits and herbal products, some of them were reported to produce transporter-mediated DDI. Our objective was to investigate potential inhibitors of OATP1B1 from 99 flavonoids, and to assess the hepatoprotective effects on bosentan induced liver injury. Eight flavonoids, including biochanin A, hispidulin, isoliquiritigenin, isosinensetin, kaempferol, licochalcone A, luteolin and sinensetin exhibited significant inhibition (>50 %) on OATP1B1 in OATP1B1-HEK293 cells, which reduced the OATP1B1-mediated influx of methotrexate, accordingly decreased its cytotoxicity in OATP1B1-HEK293 cells and increased its AUC0-t in different extents in rats, from 28.27%-82.71 %. In bosentan-induced rat liver injury models, 8 flavonoids reduced the levels of serum total bile acid (TBA) and the liver concentration of bosentan in different degrees. Among them, kaempferol decreased the concentration most significantly, by 54.17 %, which indicated that flavonoids may alleviate bosentan-induced liver injury by inhibiting OATP1B1-mediated bosentan uptake. Furthermore, the pharmacophore model indicated the hydrogen bond acceptors and hydrogen bond donors may play critical role in the potency of flavonoids inhibition on OATP1B1. Taken together, our findings would provide helpful information for predicting the potential risks of flavonoid-containing food/herb-drug interactions in humans and alleviating bosentan -induced liver injury by OATP1B1 regulation.
Collapse
Affiliation(s)
- Xiaoqing Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jie Bai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Minwan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanxia Xu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Shengyu Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanhong Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Baolian Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Yan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Beijing Key Laboratory of Active Substances Discovery and Drug Ability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
6
|
Rodrigues AD, Lai Y, Shen H, Varma MVS, Rowland A, Oswald S. Induction of Human Intestinal and Hepatic Organic Anion Transporting Polypeptides: Where Is the Evidence for Its Relevance in Drug-Drug Interactions? Drug Metab Dispos 2020; 48:205-216. [PMID: 31879282 DOI: 10.1124/dmd.119.089615] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/06/2019] [Indexed: 02/13/2025] Open
Abstract
Organic anion transporting polypeptides (OATPs), expressed in human liver (OATP1B1, OATP1B3, and OATP2B1) and intestine (OATP2B1), govern the pharmacokinetics (PK) of drugs (e.g., statins) and endogenous substrates (e.g., coproporphyrin I [CPI]). Their expression is known to be modulated (e.g., disease, age, and environmental factors), and they also present as the loci of clinically relevant polymorphisms and drug interactions involving inhibition. In comparison, relatively few clinical reports describe the induction of OATPs, although the effect of inducers (e.g., rifampicin [RIF], carbamazepine [CBZ]) on OATP biomarker plasma levels and statin PK has been reported. Of note, available human tissue (e.g., biopsy) protein and messenger RNA expression profiling data indicate that OATPs in gut and liver are not induced by prototypical inducers such as RIF when compared with cytochrome P450 3A4 (CYP3A4), P-glycoprotein (Pgp), multidrug resistance-associated protein 2 (MRP2), and breast cancer resistance protein (BCRP). Such results are consistent with in vitro human hepatocyte data. Therefore, the observed impact of RIF, and possibly CBZ, on statin PK (>20% decrease in the area under the plasma concentration vs. time curve) cannot be ascribed to OATP induction with certainty. In fact, most statins and CPI have been shown to present variously as substrates of RIF-inducible proteins such as CYP3A4, Pgp, MRP2, and BCRP. Interpretation of multidose RIF data is further complicated by its autoinduction, which likely leads to decreased inhibition of OATP. In the absence of more conclusive OATP induction data, caution is needed when modeling drug-drug interactions involving multidose inducers such as RIF. SIGNIFICANCE STATEMENT: Presently, there is limited direct clinical evidence supporting the notion that human liver and gut organic anion transporting polypeptides (OATPs) are inducible by agents like rifampicin (RIF). Such data need to be reconciled and will pose challenges for attempting to incorporate OATP induction into physiologically based pharmacokinetics models. Although disparate sets of tissue biopsy (atorvastatin and carbamazepine) and in vitro hepatocyte (phenobarbital, chenodeoxycholate, and amprenavir) data present OATP messenger RNA induction (≥2-fold) by agents beyond RIF, the clinical relevance of such data needs to be determined.
Collapse
Affiliation(s)
- A David Rodrigues
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (A.D.R., M.V.S.V.); Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (Y.L.); Department of Metabolism & Pharmacokinetics, Bristol-Myers Squibb Research & Development, Princeton, New Jersey (H.S.); College of Medicine & Public Health, Flinders University, Adelaide, South Australia, Australia (A.R.); and Department of Clinical Pharmacology, Center of Drug Absorption & Transport, University Medicine of Greifswald, Greifswald, Germany (S.O.)
| | - Yurong Lai
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (A.D.R., M.V.S.V.); Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (Y.L.); Department of Metabolism & Pharmacokinetics, Bristol-Myers Squibb Research & Development, Princeton, New Jersey (H.S.); College of Medicine & Public Health, Flinders University, Adelaide, South Australia, Australia (A.R.); and Department of Clinical Pharmacology, Center of Drug Absorption & Transport, University Medicine of Greifswald, Greifswald, Germany (S.O.)
| | - Hong Shen
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (A.D.R., M.V.S.V.); Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (Y.L.); Department of Metabolism & Pharmacokinetics, Bristol-Myers Squibb Research & Development, Princeton, New Jersey (H.S.); College of Medicine & Public Health, Flinders University, Adelaide, South Australia, Australia (A.R.); and Department of Clinical Pharmacology, Center of Drug Absorption & Transport, University Medicine of Greifswald, Greifswald, Germany (S.O.)
| | - Manthena V S Varma
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (A.D.R., M.V.S.V.); Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (Y.L.); Department of Metabolism & Pharmacokinetics, Bristol-Myers Squibb Research & Development, Princeton, New Jersey (H.S.); College of Medicine & Public Health, Flinders University, Adelaide, South Australia, Australia (A.R.); and Department of Clinical Pharmacology, Center of Drug Absorption & Transport, University Medicine of Greifswald, Greifswald, Germany (S.O.)
| | - Andrew Rowland
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (A.D.R., M.V.S.V.); Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (Y.L.); Department of Metabolism & Pharmacokinetics, Bristol-Myers Squibb Research & Development, Princeton, New Jersey (H.S.); College of Medicine & Public Health, Flinders University, Adelaide, South Australia, Australia (A.R.); and Department of Clinical Pharmacology, Center of Drug Absorption & Transport, University Medicine of Greifswald, Greifswald, Germany (S.O.)
| | - Stefan Oswald
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (A.D.R., M.V.S.V.); Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (Y.L.); Department of Metabolism & Pharmacokinetics, Bristol-Myers Squibb Research & Development, Princeton, New Jersey (H.S.); College of Medicine & Public Health, Flinders University, Adelaide, South Australia, Australia (A.R.); and Department of Clinical Pharmacology, Center of Drug Absorption & Transport, University Medicine of Greifswald, Greifswald, Germany (S.O.)
| |
Collapse
|
7
|
Niu C, Wang Y, Zhao X, Tep S, Murakami E, Subramanian R, Smith B, Lai Y. Organic Anion-Transporting Polypeptide Genes Are Not Induced by the Pregnane X Receptor Activator Rifampin: Studies in Hepatocytes In Vitro and in Monkeys In Vivo. Drug Metab Dispos 2019; 47:1433-1442. [PMID: 31582395 DOI: 10.1124/dmd.119.088922] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/27/2019] [Indexed: 12/27/2022] Open
Abstract
Induction potentials of the pregnane X receptor (PXR) activator rifampin (RIF) on transporter genes [e.g., organic anion-transporting polypeptides (OATPs)] are still in its infancy or remain controversial in the field. The present investigations characterized changes in transporter gene expression by RIF in sandwich-cultured hepatocytes from multiple donors of human and cynomolgus monkey using real-time quantitative reverse transcription polymerase chain reaction method. Three-day treatment of RIF significantly induced CYP3A4 (∼60-fold induction), but not CYP1A2 and CYP2D6 genes. SLC51B was the most highly induced uptake transporter gene (>10-fold) in both human and monkey hepatocytes. A greater induction of CYP2C9 was observed in monkey hepatocytes than that in humans. ATP-binding cassette (ABC)B1 and ABCC2 were induced slightly above 2-fold in human and monkey hepatocytes and appeared to be dose-dependent. The induction of OATP and other transporter genes was generally less than 2-fold and considered not clinically relevant. SLCO2B1 was not detectable in monkey hepatocytes. To investigate in vivo OATP induction, RIF (18 mg/kg per day) was orally dosed to cynomolgus monkeys for 7 days. Pitavastatin and antipyrine were intravenously dosed before and after RIF treatment as exogenous probes of OATP and CYP activities, respectively. Plasma coproporphyrin-I (CP-I) and coproporphyrin-III (CP-III) were measured as OATP endogenous biomarkers. Although a significant increase of antipyrine clearance (CL) was observed after RIF treatment, the plasma exposures of pitavastatin, CP-I, and CP-III remained unchanged, suggesting that OATP function was not significantly altered. The results suggested that OATP transporters were not significantly induced by PXR ligand RIF. The data are consistent with current regulatory guidances that the in vitro characterization of transporter induction during drug development is not required. SIGNIFICANCE STATEMENT: Organic anion-transporting polypeptide (OATP) genes were not induced by rifampin in sandwich-cultured human and monkey hepatocytes OATP functions measured by OATP probe pitavastatin and endogenous marker coproporphyrins were not altered in monkeys in vivo by 7-day rifampin treatment. The data suggested that OATP transporters are unlikely induced by the pregnane X receptor ligand rifampin, which are consistent with current regulatory guidances that the in vitro characterization of OATP1B induction during drug development is not required.
Collapse
Affiliation(s)
- Congrong Niu
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Yujin Wang
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Xiaofeng Zhao
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Sam Tep
- Drug Metabolism, Gilead Sciences, Foster City, California
| | | | | | - Bill Smith
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences, Foster City, California
| |
Collapse
|
8
|
Metabolic Activation of Cholestatic Drug-Induced Bile Acid-Dependent Toxicity in Human Sandwich-Cultured Hepatocytes. J Pharm Sci 2017; 106:2509-2514. [DOI: 10.1016/j.xphs.2017.04.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 11/19/2022]
|