1
|
Guo Y, Zhang H, Zhao N, Peng Y, Shen D, Chen Y, Zhang X, Tang CE, Chai J. STING-mediated IL-6 Inhibits OATP1B1 Expression via the TCF4 Signaling Pathway in Cholestasis. J Clin Transl Hepatol 2024; 12:701-712. [PMID: 39130625 PMCID: PMC11310758 DOI: 10.14218/jcth.2024.00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 08/13/2024] Open
Abstract
Background and Aims Organic anion-transporting polypeptides (OATPs) play a crucial role in the transport of bile acids and bilirubin. In our previous study, interleukin 6 (IL-6) reduced OATP1B3 levels in cholestatic disease. However, it remains unclear whether IL-6 inhibits OATP1B1 expression in cholestatic diseases. This study aimed to investigate whether IL-6 can inhibit OATP1B1 expression and explore the underlying mechanisms. Methods The effect of stimulator of interferon genes (STING) signaling on inflammatory factors was investigated in a cholestatic mouse model using RT-qPCR and enzyme-linked immunosorbent assay. To assess the impact of inflammatory factors on OATP1B1 expression in hepatocellular carcinoma, we analyzed OATP1B1 expression by RT-qPCR and Western Blot after treating PLC/PRF/5 cells with TNF-α, IL-1β, and IL-6. To elucidate the mechanism by which IL-6 inhibits OATP1B1 expression, we examined the expression of the OATP1B1 regulator TCF4 in PLC/PRF/5 and HepG2 cells using RT-qPCR and Western Blot. The interaction mechanism between β-catenin/TCF4 and OATP1B1 was investigated by knocking down β-catenin/TCF4 through siRNA transfection. Results The STING inhibitor decreased inflammatory factor levels in the cholestatic mouse model, with IL-6 exhibiting the most potent inhibitory effect on OATP1B1. IL-6 downregulated β-catenin/TCF4, leading to decreased OATP1B1 expression. Knocking-down β-catenin/TCF4 counteracted the β-catenin/TCF4-mediated repression of OATP1B1. Conclusions STING-mediated IL-6 up-regulation may inhibit OATP1B1, leading to reduced transport of bile acids and bilirubin by OATP1B1. This may contribute to altered pharmacokinetics in patients with diseases associated with increased IL-6 production.
Collapse
Affiliation(s)
- Yan Guo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Hongjia Zhang
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Nan Zhao
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Peng
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Dongya Shen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yubin Chen
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxun Zhang
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Can-E Tang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jin Chai
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
2
|
Ono H, Tanaka R, Suzuki Y, Oda A, Sato H, Tatsuta R, Ando T, Shin T, Ohno K, Itoh H. Relationship of plasma 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid concentration with OATP1B activity in patients with chronic kidney disease. Clin Transl Sci 2024; 17:e13731. [PMID: 38564661 PMCID: PMC10844757 DOI: 10.1111/cts.13731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 04/04/2024] Open
Abstract
Organic anion-transporting polypeptides (OATP)1B are drug transporters mainly expressed in the sinusoidal membrane. Many studies have suggested that OATP1B activity is affected by genetic factor, the uremic toxin 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF), and inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Coproporphyrin-I (CP-I) is spotlighted as a highly accurate endogenous substrate of OATP1B. We previously reported a positive correlation between plasma CMPF and CP-I concentrations in patients with chronic kidney disease (CKD). The present study evaluated the impact of genetic polymorphisms, CMPF, IL-6, TNF-α, and estimated glomerular filtration rate (eGFR) on individual differences in OATP1B activity in patients with CKD. Seventy-three patients with CKD who received kidney transplant at least 3 months earlier were analyzed. Plasma CP-I concentration was higher in OATP1B1*15 carriers than in non-carriers. In all patients, CP-I did not correlate significantly with CMPF, IL-6, TNF-α, or eGFR. However, when the dataset was cut off at CMPF concentration of 8 and 7 μg/mL, 4 μg/mL, 3 μg/mL or 2 μg/mL, CMPF correlated positively with CP-I, and correlation coefficient tended to be higher as plasma CMPF concentration was lower. In conclusion, OATP1B1*15 impacted OATP1B activity in patients with CKD, but IL-6 and TNF-α did not. However, the impact of CMPF on OATP1B activity was limited to low CMPF concentrations, and the effect could be saturated at high concentrations. When prescribing an OATP1B substrate drug for patients with CKD, the OATP1B1*15 carrier status and plasma CMPF concentration may need to be considered to decide the dose regimen.
Collapse
Affiliation(s)
- Hiroyuki Ono
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Ryota Tanaka
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Yosuke Suzuki
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityKiyoseTokyoJapan
| | - Ayako Oda
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityKiyoseTokyoJapan
| | - Haruki Sato
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityKiyoseTokyoJapan
| | - Ryosuke Tatsuta
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Tadasuke Ando
- Department of UrologyOita University Faculty of MedicineYufuOitaJapan
| | - Toshitaka Shin
- Department of UrologyOita University Faculty of MedicineYufuOitaJapan
| | - Keiko Ohno
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityKiyoseTokyoJapan
| | - Hiroki Itoh
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| |
Collapse
|
3
|
Song C, Kang T, Gao K, Shi X, Zhang M, Zhao L, Zhou L, Guo J. Preparation for mice spaceflight: Indications for training C57BL/6J mice to adapt to microgravity effect with three-dimensional clinostat on the ground. Heliyon 2023; 9:e19355. [PMID: 37662714 PMCID: PMC10472007 DOI: 10.1016/j.heliyon.2023.e19355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/09/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023] Open
Abstract
Like astronauts, animals need to undergo training and screening before entering space. At present, pre-launch training for mice mainly focuses on adaptation to habitat system. Training for the weightless environment of space in mice has not received much attention. Three-dimensional (3D) clinostat is a method to simulate the effects of microgravity on Earth. However, few studies have used a 3D clinostat apparatus to simulate the effects of microgravity on animal models. Therefore, we conducted a study to evaluate the feasibility and effects of long-term treatment with three-dimensional clinostat in C57BL/6 J mice. Thirty 8-week-old male C57BL/6 J mice were randomly assigned to three groups: mice in individually ventilated cages (MC group, n = 6), mice in survival boxes (SB group, n = 12), and mice in survival boxes receiving 3D clinostat treatment (CS group, n = 12). The mice showed good tolerance after 12 weeks of alternate day training. To evaluate the biological effects of simulated microgravity, the changes in serum metabolites were monitored using untargeted metabolomics, whereas bone loss was assessed using microcomputed tomography of the left femur. Compared with the metabolome of the SB group, the metabolome of the CS group showed significant differences during the first three weeks and the last three weeks. The KEGG pathways in the late stages were mainly related to the nervous system, indicating the influence of long-term microgravity on the central nervous system. Besides, a marked reduction in the trabecular number (P < 0.05) and an increasing trend of trabecular spacing (P < 0.1) were observed to occur in a time-dependent manner in the CS group compared with the SB group. These results showed that mice tolerated well in a 3D clinostat and may provide a new strategy in pre-launch training for mice and conducting relevant ground-based modeling experiments.
Collapse
Affiliation(s)
- Chenchen Song
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Taisheng Kang
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Kai Gao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xudong Shi
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Meng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Lianlian Zhao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Li Zhou
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jianguo Guo
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Mochizuki T, Kusuhara H. Progress in the Quantitative Assessment of Transporter-Mediated Drug-Drug Interactions Using Endogenous Substrates in Clinical Studies. Drug Metab Dispos 2023; 51:1105-1113. [PMID: 37169512 DOI: 10.1124/dmd.123.001285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
Variations in drug transporter activities, caused by genetic polymorphism and drug-drug interactions (DDIs), alter the systemic exposure of substrate drugs, leading to differences in drug responses. Recently, some endogenous substrates of drug transporters, particularly the solute carrier family transporters such as OATP1B1, OATP1B3, OAT1, OAT3, OCT1, OCT2, MATE1, and MATE2-K, have been identified to investigate variations in drug transporters in humans. Clinical data obtained support their performance as surrogate probes in terms of specificity and reproducibility. Pharmacokinetic parameters of the endogenous biomarkers depend on the genotypes of drug transporters and the systemic exposure to perpetrator drugs. Furthermore, the development of physiologically based pharmacokinetic models for the endogenous biomarkers has enabled a top-down approach to obtain insights into the effect of perpetrators on drug transporters and to more precisely simulate the DDI with victim drugs, including probe drugs. The endogenous biomarkers can address the uncertainty in the DDI prediction in the preclinical and early phases of clinical development and have the potential to fulfill regulatory requirements. Therefore, the endogenous biomarkers should be able to predict disease effects on the variations in drug transporter activities observed in patients. This mini-review focuses on recent progress in the identification and use of the endogenous drug transporter substrate biomarkers and their application in drug development. SIGNIFICANCE STATEMENT: Advances in analytical methods have enabled the identification of endogenous substrates of drug transporters. Changes in the pharmacokinetic parameters (Cmax, AUC, or CLR) of these endogenous biomarkers relative to baseline values can serve as a quantitative index to assess variations in drug transporter activities during clinical studies and thereby provide more precise DDI predictions.
Collapse
Affiliation(s)
- Tatsuki Mochizuki
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., Yokohama, Japan (T.M.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (H.K.)
| | - Hiroyuki Kusuhara
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., Yokohama, Japan (T.M.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan (H.K.)
| |
Collapse
|
5
|
Watanabe T, Tanaka R, Suzuki Y, Sato H, Negami J, Yoshijima C, Oda A, Ono H, Tatsuta R, Ohno K, Itoh H. Positive correlation between organic anion transporter 1B function indicated by plasma concentration of coproporphyrin-I and blood concentration of cyclosporin A in real-world patients. Br J Clin Pharmacol 2022; 89:1672-1681. [PMID: 36517987 DOI: 10.1111/bcp.15640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/20/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
AIMS Cyclosporin A (CyA) has potent inhibitory activity on organic anion transporting polypeptide 1B (OATP1B), causing drug-drug interactions with its substrate drugs. 3-carboxy-4-methyl-5-propyl-2-furanpropionate (CMPF), a uraemic toxin, has also been suggested to inhibit OATP1B activity. Recent study has identified coproporphyrin-I (CP-I) as a specific endogenous substrate for OATP1B, which is useful to indicate OATP1B activity. We investigated the relationship of CP-I with CyA and CMPF concentrations in patients taking CyA. METHODS In total, 121 blood samples from 74 patients who took CyA and underwent routine therapeutic drug monitoring were divided into trough and peak samples. RESULTS CyA and CP-I concentrations were significantly higher in peak samples than in trough samples. A positive correlation between CP-I and CyA concentrations was found in all samples and in trough and peak samples, while no correlation was observed between CP-I and CMPF concentrations. Multiple regression analysis identified CyA and C-reactive protein concentrations as independent factors affecting CP-I concentration, with blood CyA concentration having markedly greater contribution to plasma CP-I concentration. CONCLUSION The present study suggests that CyA inhibits OATP1B activity in a concentration-dependent manner in clinical setting, and that dose adjustment of OATP1B substrate drugs coadministered with CyA according to plasma CMPF concentration may not be necessary.
Collapse
Affiliation(s)
- Takuma Watanabe
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Ryota Tanaka
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Yosuke Suzuki
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Haruki Sato
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Jun Negami
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Chisato Yoshijima
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Ayako Oda
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Hiroyuki Ono
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Ryosuke Tatsuta
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Keiko Ohno
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Hiroki Itoh
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| |
Collapse
|
6
|
Fujino C, Ueshima S, Katsura T. Changes in the expression of drug-metabolising enzymes and drug transporters in mice with collagen antibody-induced arthritis. Xenobiotica 2022; 52:758-766. [PMID: 36278306 DOI: 10.1080/00498254.2022.2137442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
1. We investigated the changes in the expression of drug-metabolising enzymes and drug transporters in the liver, small intestine and kidney of mice with collagen antibody-induced arthritis (CAIA) to determine whether changes in these expressions affect pharmacokinetics of drugs in patients with rheumatoid arthritis.2. mRNA expression levels of cytochrome P450 (Cyp) 2b10, Cyp2c29 and Cyp3a11 were observed to be lower in the liver and small intestine of CAIA mice than in control mice. Compared with control mice, mRNA expression levels of multidrug resistance 1 b, peptide transporter 2 and organic anion transporter (Oat) 2 were high in the liver of CAIA mice. Changes in these expression levels were different among organs. However, elevated expression of Oat2 mRNA was not associated with an increase in protein expression and transport activity evaluated using [3H]cGMP as a substrate.3. These results suggest that arthritis can change the expression of pharmacokinetics-related genes, but the changes may not necessarily be linked to the pharmacokinetics in patients with rheumatoid arthritis. On the other hand, we found Oat2 mRNA expression level was positively correlated with plasma interleukin-6 level, indicating that transcriptional activation of Oat2 may occur in inflammatory state.
Collapse
Affiliation(s)
- Chieri Fujino
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Satoshi Ueshima
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Toshiya Katsura
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| |
Collapse
|
7
|
Chu X, Prasad B, Neuhoff S, Yoshida K, Leeder JS, Mukherjee D, Taskar K, Varma MVS, Zhang X, Yang X, Galetin A. Clinical Implications of Altered Drug Transporter Abundance/Function and PBPK Modeling in Specific Populations: An ITC Perspective. Clin Pharmacol Ther 2022; 112:501-526. [PMID: 35561140 DOI: 10.1002/cpt.2643] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022]
Abstract
The role of membrane transporters on pharmacokinetics (PKs), drug-drug interactions (DDIs), pharmacodynamics (PDs), and toxicity of drugs has been broadly recognized. However, our knowledge of modulation of transporter expression and/or function in the diseased patient population or specific populations, such as pediatrics or pregnancy, is still emerging. This white paper highlights recent advances in studying the changes in transporter expression and activity in various diseases (i.e., renal and hepatic impairment and cancer) and some specific populations (i.e., pediatrics and pregnancy) with the focus on clinical implications. Proposed alterations in transporter abundance and/or activity in diseased and specific populations are based on (i) quantitative transporter proteomic data and relative abundance in specific populations vs. healthy adults, (ii) clinical PKs, and emerging transporter biomarker and/or pharmacogenomic data, and (iii) physiologically-based pharmacokinetic modeling and simulation. The potential for altered PK, PD, and toxicity in these populations needs to be considered for drugs and their active metabolites in which transporter-mediated uptake/efflux is a major contributor to their absorption, distribution, and elimination pathways and/or associated DDI risk. In addition to best practices, this white paper discusses current challenges and knowledge gaps to study and quantitatively predict the effects of modulation in transporter activity in these populations, together with the perspectives from the International Transporter Consortium (ITC) on future directions.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Department of ADME and Discovery Toxicology, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | | | - Kenta Yoshida
- Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, California, USA
| | - James Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Dwaipayan Mukherjee
- Clinical Pharmacology & Pharmacometrics, Research & Development, AbbVie, Inc., North Chicago, Illinois, USA
| | | | - Manthena V S Varma
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut, USA
| | - Xinyuan Zhang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xinning Yang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Mochizuki T, Aoki Y, Yoshikado T, Yoshida K, Lai Y, Hirabayashi H, Yamaura Y, Rockich K, Taskar K, Takashima T, Chu X, Zamek-Gliszczynski MJ, Mao J, Maeda K, Furihata K, Sugiyama Y, Kusuhara H. Physiologically-based pharmacokinetic model-based translation of OATP1B-mediated drug-drug interactions from coproporphyrin I to probe drugs. Clin Transl Sci 2022; 15:1519-1531. [PMID: 35421902 DOI: 10.1111/cts.13272] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 11/28/2022] Open
Abstract
The accurate prediction of OATP1B-mediated drug-drug interactions (DDIs) is challenging for drug development. Here, we report physiologically-based pharmacokinetic (PBPK) model analysis for clinical DDI data generated in heathy subjects who received oral doses of cyclosporin A (CysA; 20 and 75 mg) as an OATP1B inhibitor, and the probe drugs (pitavastatin, rosuvastatin and valsartan). PBPK models of CysA and probe compounds were combined assuming inhibition of hepatic uptake of endogenous coproporphyrin I (CP-I) by CysA. In vivo Ki of unbound CysA for OATP1B (Ki,OATP1B ), and the overall intrinsic hepatic clearance per body weight of CP-I (CLint,all,unit ) were optimized to account for the CP-I data (Ki,OATP1B , 0.657 ± 0.048 nM; CLint,all,unit , 57.0 ± 6.3 L/h/kg). DDI simulation using Ki,OATP1B reproduced the dose-dependent effect of CysA (20 and 75 mg) and the dosing interval (1 h and 3 h) on the time profiles of blood concentrations of pitavastatin and rosuvastatin, but DDI simulation using in vitro Ki,OATP1B failed. The Cluster Gauss-Newton method was used to conduct parameter optimization using 1,000 initial parameter sets for the seven pharmacokinetic parameters of CP-I (β, CLint,all , Fa Fg , Rdif , fbile , fsyn , and vsyn ), and Ki,OATP1B , and Ki,MRP2 of CysA. Based on the accepted 498 parameter sets, the range of CLint,all and Ki,OATP1B was narrowed, with coefficients of variation (CVs) of 9.3% and 11.1%, respectively, indicating that these parameters were practically identifiable. These results suggest that PBPK model analysis of CP-I is a promising translational approach to predict OATP1B-mediated DDIs in drug development.
Collapse
Affiliation(s)
- Tatsuki Mochizuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo
| | - Yasunori Aoki
- Laboratory of quantitative system pharmacokinetics / pharmacodynamics, Josai International University, School of Pharmacy, Tokyo, Japan
| | - Takashi Yoshikado
- Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Yokohama, Kanagawa, Japan
| | - Kenta Yoshida
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California, USA
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Yoshiyuki Yamaura
- Pharmacokinetic Research Laboratories , Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Kevin Rockich
- Drug Metabolism, Pharmacokinetics and Clinical Pharmacology, Incyte Research Institute, Wilmington, Delaware, USA
| | - Kunal Taskar
- Drug Metabolism and Pharmacokinetics, IVIVT, GlaxoSmithKline, Stevenage, UK
| | - Tadayuki Takashima
- Laboratory for Safety Assessment & ADME, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Jialin Mao
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo
| | | | - Yuichi Sugiyama
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo.,Laboratory of quantitative system pharmacokinetics / pharmacodynamics, Josai International University, School of Pharmacy, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo
| |
Collapse
|