1
|
Kosicka-Noworzyń K, Romaniuk-Drapała A, Sheng YH, Yohn C, Brunetti L, Kagan L. Obesity-related drug-metabolizing enzyme expression alterations in the human liver. Biomed Pharmacother 2025; 187:118155. [PMID: 40359692 DOI: 10.1016/j.biopha.2025.118155] [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: 01/14/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE Implications of obesity extend beyond the association with various health conditions, impacting physiological changes that affect the liver and the activity of metabolizing enzymes. Given the prevalence of obesity and the risk for drug-drug interactions owing to the comorbidity burden, the current drug dosage recommendations may need reevaluation for patients with obesity. This study evaluated the implications of obesity on the gene expression of hepatic drug-metabolizing enzymes. As drug clearance is an essential pharmacokinetic parameter for maintaining drug dosing regimens, investigating alterations in metabolizing enzymes expression is a critical step. METHODS Human liver samples were collected post-mortem from 32 individuals and classified into the control (18.5 ≤ BMI <25 kg/m2; range 18.9-24.4 kg/m2; median 22.3 kg/m2) and the study group (BMI ≥25 kg/m2; range 25.1-55.5 kg/m2; median 31.2 kg/m2). Real-time quantitative PCR was performed for the analysis of 168 drug-metabolizing enzymes. RESULTS Our studies revealed several potential physiologically relevant differences, but the statistical significance was reached only for ALDH3B1, PTGS1, and CEL (all being up-regulated in the study group). CONCLUSIONS The study adds to our understanding of the mechanisms of pharmacokinetic changes in overweight and obesity. The findings require further exploration on the protein level, through proteomic and functional studies.
Collapse
Affiliation(s)
- Katarzyna Kosicka-Noworzyń
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3, Poznań 60-806, Poland; Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA.
| | - Aleksandra Romaniuk-Drapała
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3, Poznań 60-806, Poland; Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA.
| | - Yi-Hua Sheng
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA.
| | - Christine Yohn
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA.
| | - Luigi Brunetti
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA.
| | - Leonid Kagan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA; Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
2
|
Yang C, Gong J, Xue M, Huang W, Yuan Y, Chen C, He Y, Yang C, Sun H, Liu Y, Gong Y, Wu Y, Lai X, Zhong D, Diao X, Lu H, Zheng Y. Effects of the Thiol Methyltransferase Inhibitor (±)-2,3-Dichloro- α-Methylbenzylamine (DCMB) on the Pharmacokinetics and Metabolism of Vicagrel in Rats. Drug Metab Dispos 2024; 52:988-996. [PMID: 38997155 DOI: 10.1124/dmd.124.001739] [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: 04/01/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
P2Y12 receptor inhibitors are commonly used in clinical antiplatelet therapy, typically alongside other medications. Vicagrel, a promising P2Y12 receptor inhibitor, has submitted a new drug marketing application to the United States Food and Drug Administration. Its primary metabolites and some metabolic pathways are identical to those of clopidogrel. The aim of this study was to investigate the effects of the thiol methyltransferase inhibitor (±)-2,3-dichloro-α-methylbenzylamine (DCMB) on the metabolism and pharmacokinetics of vicagrel. In vitro incubation with human and rat liver microsomes revealed that DCMB significantly inhibited the methylation of vicagrel's thiol metabolite M15-1. Rats were orally administered 6 mg/kg [14C]vicagrel (100 μCi/kg) 1 hour after peritoneal injection with or without DCMB (80 mg/kg). Compared with the control group, the plasma of DCMB-pretreated rats exhibited maximum plasma concentration (C max) decrease and time to reach C max (T max) delay for all vicagrel-related substances, the methylation product of the thiol metabolite (M9-2), and the derivatization product of the active thiol metabolite (MP-M15-2). However, no significant changes in area under the curve (AUC) or half-life (t 1/2) were observed. DCMB had negligible effect on the total radiological recovery of vicagrel within 72 hours, although the rate of vicagrel excretion slowed down within 48 hours. DCMB had a negligible impact on the metabolic pathway of vicagrel. Overall, the present study found that DCMB did not significantly affect the total exposure, metabolic pathways, metabolite profiles, or total excretion rates of vicagrel-related metabolites in rats, but led to C max decrease, T max delay, and slower excretion rate within 48 hours. SIGNIFICANCE STATEMENT: This study used liquid chromatography-tandem mass spectrometry combined with radiolabeling technology to investigate the effects of the thiol methyltransferase inhibitor (±)-2,3-dichloro-α-methylbenzylamine on the absorption, metabolism, and excretion of vicagrel in rats. This work helps to better understand the in vivo metabolism of active thiol metabolites of P2Y12 inhibitors such as clopidogrel, vicagrel, etc.
Collapse
Affiliation(s)
- Cheng Yang
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Jingru Gong
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Mingzhen Xue
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Wensi Huang
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Yali Yuan
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Chong Chen
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Yifei He
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Chen Yang
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Hongbin Sun
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Yongqiang Liu
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Yanchun Gong
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Yong Wu
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Xiaojuan Lai
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Dafang Zhong
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Xingxing Diao
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Huiping Lu
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| | - Yuandong Zheng
- Shanghai Center for Drug Metabolism and Pharmacokinetics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (M.X., W.H., Y.Y., C.C., Y.H., Chen Y., D.Z., X.D., Y.Z.); School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China (Cheng Y., Y.Y., C.C., X.D.); Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (J.G., H.L.); Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China (M.X.); State Key Laboratory of Natural Medicines and Center of Drug Discovery, College of Pharmacy, China Pharmaceutical University, Nanjing, China (H.S.); and Jiangsu Vcare PharmaTech Co. Ltd., Nanjing, China (Y.L., Y.G., Y.W., X.L.)
| |
Collapse
|
3
|
Kamp AL, Yousofzai W, Kooistra HS, Santarelli G, van Geijlswijk IM. Suspected clopidogrel-associated hepatitis in a cat. JFMS Open Rep 2024; 10:20551169241278408. [PMID: 39512443 PMCID: PMC11542135 DOI: 10.1177/20551169241278408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Case summary Acute hepatitis and liver damage are rare adverse effects of clopidogrel in humans. In veterinary medicine, clopidogrel is mainly prescribed in the treatment of feline patients with cardiomyopathies. Little is known regarding the safety and adverse effects of clopidogrel in this group of patients. The limited number of studies scarcely report adverse effects. In this case report, a 6-year-old male castrated crossbred cat with the hypertrophic cardiomyopathy phenotype had signs of acute hepatitis after 5 weeks of clopidogrel treatment. Relevance and novel information Evaluation of the case and review of the literature indicate that acute hepatitis might be a potential adverse effect of clopidogrel in feline patients. Therefore, hepatotoxicity should be taken into consideration when a feline patient shows clinical deterioration after the use of clopidogrel.
Collapse
Affiliation(s)
- Astrid L Kamp
- Department Clinical Sciences, Division Internal Medicine of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Washma Yousofzai
- Department Population Health Sciences, IRAS Veterinary and Comparative Pharmacology Group (One Health Pharma) – Pharmacy, Utrecht University, Utrecht, The Netherlands
| | - Hans S Kooistra
- Department Clinical Sciences, Division Internal Medicine of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Giorgia Santarelli
- Department Clinical Sciences, Division Internal Medicine of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Ingeborg M van Geijlswijk
- Department Population Health Sciences, IRAS Veterinary and Comparative Pharmacology Group (One Health Pharma) – Pharmacy, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
4
|
Jmel H, Boukhalfa W, Gouiza I, Seghaier RO, Dallali H, Kefi R. Pharmacogenetic landscape of pain management variants among Mediterranean populations. Front Pharmacol 2024; 15:1380613. [PMID: 38813106 PMCID: PMC11134176 DOI: 10.3389/fphar.2024.1380613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/05/2024] [Indexed: 05/31/2024] Open
Abstract
Background Chronic pain is a major socioeconomic burden in the Mediterranean region. However, we noticed an under-representation of these populations in the pharmacogenetics of pain management studies. In this context, we aimed 1) to decipher the pharmacogenetic variant landscape among Mediterranean populations compared to worldwide populations in order to identify therapeutic biomarkers for personalized pain management and 2) to better understand the biological process of pain management through in silico investigation of pharmacogenes pathways. Materials and Methods We collected genes and variants implicated in pain response using the Prisma guidelines from literature and PharmGK database. Next, we extracted these genes from genotyping data of 829 individuals. Then, we determined the variant distribution among the studied populations using multivariate (MDS) and admixture analysis with R and STRUCTURE software. We conducted a Chi2 test to compare the interethnic frequencies of the identified variants. We used SNPinfo web server, miRdSNP database to identify miRNA-binding sites. In addition, we investigated the functions of the identified genes and variants using pathway enrichment analysis and annotation tools. Finally, we performed docking analysis to assess the impact of variations on drug interactions. Results We identified 63 variants implicated in pain management. MDS analysis revealed that Mediterranean populations are genetically similar to Mexican populations and divergent from other populations. STRUCTURE analysis showed that Mediterranean populations are mainly composed of European ancestry. We highlighted differences in the minor allele frequencies of three variants (rs633, rs4680, and rs165728) located in the COMT gene. Moreover, variant annotation revealed ten variants with potential miRNA-binding sites. Finally, protein structure and docking analysis revealed that two missense variants (rs4680 and rs6267) induced a decrease in COMT protein activity and affinity for dopamine. Conclusion Our findings revealed that Mediterranean populations diverge from other ethnic groups. Furthermore, we emphasize the importance of pain-related pathways and miRNAs to better implement these markers as predictors of analgesic responses in the Mediterranean region.
Collapse
Affiliation(s)
- Haifa Jmel
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Genetic Typing Service, Institut Pasteur de Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
| | - Wided Boukhalfa
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Genetic Typing Service, Institut Pasteur de Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis, Tunisia
| | - Ismail Gouiza
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Genetic Typing Service, Institut Pasteur de Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis, Tunisia
- MitoLab Team, Unité MitoVasc, Unité Mixte de Recherche Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale U1083, SFR ICAT, University of Angers, Angers, France
| | - Roua Ouled Seghaier
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Hamza Dallali
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Genetic Typing Service, Institut Pasteur de Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
| | - Rym Kefi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur de Tunis, Tunis, Tunisia
- Genetic Typing Service, Institut Pasteur de Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
5
|
Ahire D, Mariasoosai C, Naji-Talakar S, Natesan S, Prasad B. Promiscuity and Quantitative Contribution of UGT2B17 in Drug and Steroid Metabolism Determined by Experimental and Computational Approaches. J Chem Inf Model 2024; 64:483-498. [PMID: 38198666 DOI: 10.1021/acs.jcim.3c01514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Uridine 5'-diphospho-glulcuronosyltransferase 2B17 (UGT2B17) is important in the metabolism of steroids and orally administered drugs due to its high interindividual variability. However, the structural basis governing the substrate selectivity or inhibition of UGT2B17 remains poorly understood. This study investigated 76 FDA-approved drugs and 20 steroids known to undergo glucuronidation for their metabolism by UGT2B17. Specifically, we assessed the substrate selectivity for UGT2B17 over other UGT enzymes using recombinant human UGT2B17 (rUGT2B17), human intestinal microsomes, and human liver microsomes. The quantitative contribution of intestinal UGT2B17 in the glucuronidation of these compounds was characterized using intestinal microsomes isolated from UGT2B17 expressors and nonexpressors. In addition, a structure-based pharmacophore model for UGT2B17 substrates was built and validated using the studied pool of substrates and nonsubstrates. The results show that UGT2B17 could metabolize 23 out of 96 compounds from various chemical classes, including alcohols and carboxylic acids, particularly in the intestine. Interestingly, amines were less susceptible to UGT2B17 metabolism, though they could inhibit the enzyme. Three main pharmacophoric features of UGT2B17 substrates include (1) the presence of an accessible -OH or -COOH group near His35 residue, (2) a hydrophobic functional group at ∼4.5-5 Å from feature 1, and (3) an aromatic ring ∼5-7 Å from feature 2. Most of the studied compounds inhibited UGT2B17 activity irrespective of their substrate potential, indicating the possibility of multiple mechanisms. These data suggest that UGT2B17 is promiscuous in substrate selectivity and inhibition and has a high potential to produce significant variability in the absorption and disposition of orally administered drugs.
Collapse
Affiliation(s)
- Deepak Ahire
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Charles Mariasoosai
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Siavosh Naji-Talakar
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Senthil Natesan
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| |
Collapse
|
6
|
Iga K, Kiriyama A. Interplay of UDP-Glucuronosyltransferase and CYP2C8 for CYP2C8 Mediated Drug Oxidation and Its Impact on Drug-Drug Interaction Produced by Standardized CYP2C8 Inhibitors, Clopidogrel and Gemfibrozil. Clin Pharmacokinet 2024; 63:43-56. [PMID: 37921907 DOI: 10.1007/s40262-023-01322-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Early investigations into drug-drug interactions (DDIs) involving cytochrome P450 2C8 (CYP2C8) have highlighted the complexity of interactions between CYP2C8 substrate drugs, including montelukast, desloratadine, pioglitazone, repaglinide, and cerivastatin (the latter two being OATP1B1 substrates), and standardized CYP2C8 inhibitors such as clopidogrel (Clop) and gemfibrozil (Gem). These interactions have proven challenging to predict based solely on simple CYP inhibition. A hypothesis has emerged suggesting that these substrate drugs first distribute to UDP-glucuronosyltransferase (UGT) before undergoing oxidation by CYP2C8, resulting in bidirectional elimination. The process of drug distribution to UGT is believed to significantly impact these DDIs. This study aims to explore the intricate interplay between UGT and CYP2C8 in the context of DDIs involving CYP2C8 substrates affected by Clop and Gem. METHODS Plasma-level data for the unchanged drug and its metabolite, drawn from the respective literature, formed the basis of our analysis. We evaluated the enzymatic inhibitory activities of DDIs and utilized simulations to estimate plasma levels of the unchanged victim drug and its metabolite in each DDI. This was accomplished by employing a functional relationship that considered the fractional contributions of CYP2C8 and UGT to clearance, perpetrator-specific inhibitory activities against CYP2C8, and drug distribution to UGT. RESULTS Our findings emphasize the pivotal role of UGT-mediated distribution in the context of CYP2C8 substrate metabolism, particularly in the complex DDIs induced by Clop and Gem. In these DDIs, Gem exerts inhibitory effects on both UGT and CYP2C8, whereas Clop (specifically its metabolite, Clop-COOH) solely targets CYP2C8. Importantly, the inhibition of CYP2C8 by both Clop and Gem is achieved through a non-competitive mechanism, driven by the actions of their acyl-glucuronides. Clop and Gem exhibit inhibition activities accounting for 85% (pAi,CYP2C8 = 7) and 93% (pAi,CYP2C8 = 15), respectively. In contrast, Gem's inhibition of UGT is relatively modest (50%, pAi,UGT(d) = 2), and it operates through a non-specific, competitive process in drug distribution to UGT. Within this context, our UGT-CYP2C8 interplay model offers an accurate means of predicting the alterations resulting from DDIs, encompassing changes in plasma levels of the unchanged drug and its metabolites, as well as shifts in metabolite formation rates. Our analysis highlights the critical importance of considering the fractional contributions of CYP2C8 and UGT to the victim drug's clearance (fm,CYP2C8; fm,UGT) in DDI prediction. Furthermore, our examination of DDIs involving OATP1B1 substrate drugs underscores that accounting for the hepatic uptake transporters' role in the liver is superfluous in DDI prediction. CONCLUSION These findings substantially enhance our comprehension of CYP2C8-mediated oxidation and DDIs, holding crucial implications for drug development and the planning of clinical trials involving these inhibitors.
Collapse
Affiliation(s)
- Katsumi Iga
- Pharmaceutical Research and Technology Unit, R & D Division, Pre-formulation Department, Towa Pharmaceutical Co., Ltd, Kyoto Research Park KISTIC #202, 134, Chudoji Minami-machi, Shimogyo-ku, Kyoto, 600-8813, Japan.
| | - Akiko Kiriyama
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kodo Kyotanabe-shi, Kyoto, 610-0395, Japan
| |
Collapse
|
7
|
Inhibition of CYP2C8 by Acyl Glucuronides of Gemfibrozil and Clopidogrel: Pharmacological Significance, Progress and Challenges. Biomolecules 2022; 12:biom12091218. [PMID: 36139056 PMCID: PMC9496539 DOI: 10.3390/biom12091218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
The lipid-regulating drug gemfibrozil is a useful medication for reducing high cholesterol and triglycerides in the blood. In addition to oxidation, it undergoes extensive glucuronidation to produce gemfibrozil acyl glucuronide, which is a known mechanism-based inactivator of cytochrome P450 (CYP) 2C8. Such selective and time-dependent inhibition results in clinically important drug–drug interactions (DDI) with the drugs metabolized by CYP2C8. Similarly, the acyl glucuronide of clopidogrel, a widely used antiplatelet agent, is a potent time-dependent inhibitor of CYP2C8 that demonstrated significant DDI with the substrates of CYP2C8. Current progress in atomic-level understanding mostly involves studying how different drugs bind and undergo oxidation in the active site of CYPs. It is not clear how an acyl glucuronide metabolite of the drug gemfibrozil or clopidogrel interacts in the active site of CYP2C8 and selectively inhibit the enzyme. This mini-review summarizes the current knowledge on some of the important clinical DDI caused by gemfibrozil and clopidogrel due to the inhibition of CYP2C8 by acyl glucuronide metabolites of these drugs. Importantly, it examines recent developments and potential applications of structural biology tools to elucidate the binding and orientation of gemfibrozil acyl glucuronide and clopidogrel acyl glucuronide in the active site near heme that contributes to the inhibition and inactivation of CYP2C8.
Collapse
|
8
|
Physiologically Based Pharmacokinetic (PBPK) Modeling of Clopidogrel and Its Four Relevant Metabolites for CYP2B6, CYP2C8, CYP2C19, and CYP3A4 Drug–Drug–Gene Interaction Predictions. Pharmaceutics 2022; 14:pharmaceutics14050915. [PMID: 35631502 PMCID: PMC9145019 DOI: 10.3390/pharmaceutics14050915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
The antiplatelet agent clopidogrel is listed by the FDA as a strong clinical index inhibitor of cytochrome P450 (CYP) 2C8 and weak clinical inhibitor of CYP2B6. Moreover, clopidogrel is a substrate of—among others—CYP2C19 and CYP3A4. This work presents the development of a whole-body physiologically based pharmacokinetic (PBPK) model of clopidogrel including the relevant metabolites, clopidogrel carboxylic acid, clopidogrel acyl glucuronide, 2-oxo-clopidogrel, and the active thiol metabolite, with subsequent application for drug–gene interaction (DGI) and drug–drug interaction (DDI) predictions. Model building was performed in PK-Sim® using 66 plasma concentration-time profiles of clopidogrel and its metabolites. The comprehensive parent-metabolite model covers biotransformation via carboxylesterase (CES) 1, CES2, CYP2C19, CYP3A4, and uridine 5′-diphospho-glucuronosyltransferase 2B7. Moreover, CYP2C19 was incorporated for normal, intermediate, and poor metabolizer phenotypes. Good predictive performance of the model was demonstrated for the DGI involving CYP2C19, with 17/19 predicted DGI AUClast and 19/19 predicted DGI Cmax ratios within 2-fold of their observed values. Furthermore, DDIs involving bupropion, omeprazole, montelukast, pioglitazone, repaglinide, and rifampicin showed 13/13 predicted DDI AUClast and 13/13 predicted DDI Cmax ratios within 2-fold of their observed ratios. After publication, the model will be made publicly accessible in the Open Systems Pharmacology repository.
Collapse
|
9
|
Duong JK, Nand RA, Patel A, Della Pasqua O, Gross AS. A physiologically based pharmacokinetic model of clopidogrel in populations of European and Japanese ancestry: An evaluation of CYP2C19 activity. Pharmacol Res Perspect 2022; 10:e00946. [PMID: 35307978 PMCID: PMC8934724 DOI: 10.1002/prp2.946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/21/2022] Open
Abstract
Treatment response to clopidogrel is associated with CYP2C19 activity through the formation of the active H4 metabolite. The aims of this study were to develop a physiologically based pharmacokinetic (PBPK) model of clopidogrel and its metabolites for populations of European ancestry, to predict the pharmacokinetics in the Japanese population by CYP2C19 phenotype, and to investigate the effect of clinical and demographic factors. A PBPK model was developed and verified to describe the two metabolic pathways of clopidogrel (H4 metabolite, acyl glucuronide metabolite) for a population of European ancestry using plasma data from published studies. Subsequently, model predictions in the Japanese population were evaluated. The effects of CYP2C19 activity, fluvoxamine coadministration (CYP2C19 inhibitor), and population-specific factors (age, sex, BMI, body weight, cancer, hepatic, and renal dysfunction) on the pharmacokinetics of clopidogrel and its metabolites were then characterized. The predicted/observed ratios for clopidogrel and metabolite exposure parameters were acceptable (twofold acceptance criteria). For all CYP2C19 phenotypes, steady-state AUC0-τ of the H4 metabolite was lower for the Japanese (e.g., EM, 7.69 [6.26-9.45] ng·h/ml; geometric mean [95% CI]) than European (EM, 24.8 [20.4-30.1] ng·h/ml, p < .001) population. In addition to CYP2C19-poor metabolizer phenotype, fluvoxamine coadministration, hepatic, and renal dysfunction were found to reduce H4 metabolite but not acyl glucuronide metabolite concentrations. This is the first PBPK model describing the two major metabolic pathways of clopidogrel, which can be applied to populations of European and Japanese ancestry by CYP2C19 phenotype. The differences between the two populations appear to be determined primarily by the effect of varying CYP2C19 liver activity.
Collapse
Affiliation(s)
- Janna K. Duong
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DErmingtonAustralia
| | - Romina A. Nand
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DErmingtonAustralia
| | - Aarti Patel
- Drug Metabolism and PharmacokineticsGlaxoSmithKline R&DStevenageUK
| | - Oscar Della Pasqua
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DBrentfordUK
| | - Annette S. Gross
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DErmingtonAustralia
| |
Collapse
|
10
|
Zhang H, Wolford C, Basit A, Li AP, Fan PW, Murray BP, Takahashi RH, Khojasteh SC, Smith BJ, Thummel KE, Prasad B. Regional Proteomic Quantification of Clinically Relevant Non-Cytochrome P450 Enzymes along the Human Small Intestine. Drug Metab Dispos 2020; 48:528-536. [PMID: 32350063 DOI: 10.1124/dmd.120.090738] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/18/2020] [Indexed: 02/13/2025] Open
Abstract
Current challenges in accurately predicting intestinal metabolism arise from the complex nature of the intestine, leading to limited applicability of available in vitro tools as well as knowledge deficits in intestinal physiology, including enzyme abundance. In particular, information on regional enzyme abundance along the small intestine is lacking, especially for non-cytochrome P450 enzymes such as carboxylesterases (CESs), UDP-glucuronosyltransferases (UGTs), and sulfotransferases (SULTs). We used cryopreserved human intestinal mucosa samples from nine donors as an in vitro surrogate model for the small intestine and performed liquid chromatography tandem mass spectrometry-based quantitative proteomics for 17 non-cytochrome P450 enzymes using stable isotope-labeled peptides. Relative protein quantification was done by normalization with enterocyte marker proteins, i.e., villin-1, sucrase isomaltase, and fatty acid binding protein 2, and absolute protein quantification is reported as picomoles per milligram of protein. Activity assays in glucuronidations and sequential metabolisms were conducted to validate the proteomics findings. Relative or absolute quantifications are reported for CES1, CES2, five UGTs, and four SULTs along the small intestine: duodenum, jejunum, and ileum for six donors and in 10 segments along the entire small intestine (A-J) for three donors. Relative quantification using marker proteins may be beneficial in further controlling for technical variabilities. Absolute quantification data will allow for scaling factor generation and in vivo extrapolation of intestinal clearance using physiologically based pharmacokinetic modeling. SIGNIFICANCE STATEMENT: Current knowledge gaps exist in intestinal protein abundance of non-cytochrome P450 enzymes. Here, we employ quantitative proteomics to measure non-cytochrome P450 enzymes along the human small intestine in nine donors using cryopreserved human intestinal mucosa samples. Absolute and relative abundances reported here will allow better scaling of intestinal clearance.
Collapse
Affiliation(s)
- Haeyoung Zhang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Chris Wolford
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Abdul Basit
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Albert P Li
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Peter W Fan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Bernard P Murray
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Ryan H Takahashi
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - S Cyrus Khojasteh
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Bill J Smith
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Kenneth E Thummel
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington (H.Z., C.W., A.B., K.E.T., B.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (A.B., B.P.); In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L.); Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Boston, Massachusetts (P.W.F.); Genentech Inc., South San Francisco, California (R.H.T., S.C.K.); and Drug Metabolism Department, Gilead Sciences Inc., Foster City, California (B.J.S., B.P.M.)
| |
Collapse
|
11
|
Pei T, Yang J, Hu C, Chen X, Gong S, Hu X, Li L, Zhang L. Pharmacokinetics and Bioequivalence of Clopidogrel Hydrogen Sulfate Tablets in Fed and Fasted Conditions: An Open-Label, Randomized, Semireplicated Crossover Study in Healthy Chinese Volunteers. Clin Pharmacol Drug Dev 2020; 9:813-820. [PMID: 32452659 DOI: 10.1002/cpdd.804] [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: 10/23/2019] [Accepted: 03/07/2020] [Indexed: 11/11/2022]
Abstract
Clopidogrel is an antiplatelet drug with high intraindividual variability in systemic exposure and efficacy. It has been used for treating atherosclerosis and acute coronary syndrome and in preventing stent restenosis and thrombotic complications after stent implantation in clinical practice for nearly 20 years. In this study we aimed to evaluate the bioequivalence of 2 clopidogrel hydrogen sulfate formulations (75-mg tablets) under fed (n = 66) and fasted (n = 66) conditions by using the reference-scaled average bioequivalence method. An open-label, randomized, 3-sequence and 3-period crossover (3×3), semireplicated study was designed and conducted. Clopidogrel concentration of plasma samples was measured by high-precision liquid chromatography and tandem mass spectrometry. The pharmacokinetic parameters were derived by a noncompartmental model. In the fed condition the geometric least-squares mean ratios of peak concentration (Cmax ) and area under the concentration-time curve (AUC0-t ) were, respectively, 103.38% and 98.97%, and the corresponding 90%CIs were 95.68% to 111.70% and 94.67% to 103.47%. In the fasted condition the geometric least squares mean ratios of Cmax and AUC0-t were, respectively, 106.53% and 105.77%, and the corresponding 90%CIs were 97.62% to 116.25% and 96.96% to 115.38%. According to the criteria for bioequivalence (80.00% to 125.00%), the test formulations of clopidogrel and Plavix were determined to be bioequivalent.
Collapse
Affiliation(s)
- Tong Pei
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Jing Yang
- Department of Cardiology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Chaoying Hu
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Xiaoping Chen
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Shili Gong
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Xiao Hu
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Lin Li
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Phase I Clinical Trial Center, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Chen H, Jiao L, Zhou J, Bai H, Lyu M, Wu T, Wu L, Song J, Liu T, Yan H, Ying B. Absence of significant association between UGT2B4 genetic variants and the susceptibility to anti-tuberculosis drug-induced liver injury in a Western Chinese population. J Clin Pharm Ther 2020; 46:66-73. [PMID: 32170986 DOI: 10.1111/jcpt.13132] [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: 10/26/2019] [Revised: 01/11/2020] [Accepted: 02/24/2020] [Indexed: 02/05/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Combination regimens of six-month duration may increase the incidence of anti-tuberculosis drug-induced liver injury (ATLI), which is clinically characterized by mild cholestasis and hepatocanalicular lesions. UGT2B4 is a predominant UDP-glucuronosyltransferase enzyme in the human liver that plays an important role in the detoxification of bile acids, which yields water-soluble inactive compounds that can easily be excreted in the bile or urine. This study aimed to investigate the potential association between UGT2B4 variants and the susceptibility to ATLI. METHODS Genomic DNA was extracted from whole blood sample of each patient, and all SNPs were genotyped using an improved multiplex ligation detection reaction method. Clinical symptoms and laboratory results were recorded regularly. Five genetic variants at UGT2B4(rs1131878, rs1966151, rs28361541, rs4557343 and rs79407331) were identified in a prospective study of 118 ATLI cases and 628 non-ATLI controls. All participants were treated by first-line anti-TB drugs in Western China Hospital. The potential association between SNPs, ATLI risk and clinical phenotypes were determined based on the distribution of allelic frequencies and different genetic models. RESULTS AND DISCUSSION Statistical comparisons of cases and controls after correction for multiple testing did not yield any significant association between genetic variants at UGT2B4 and risk of ATLI via the analyses of single locus and subgroup differences. WHAT IS NEW AND CONCLUSION This is the first study aimed to investigate the association of UGT2B4 polymorphisms with ATLI risk. Our results revealed that UGT2B4 genetic variants are unlikely to confer susceptibility to ATLI in the Western Chinese Han population.
Collapse
Affiliation(s)
- Hao Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Jiao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Mengyuan Lyu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lijuan Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiajia Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tangyuheng Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Yan
- Department of Laboratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Zhang H, Basit A, Wolford C, Chen KF, Gaedigk A, Lin YS, Leeder JS, Prasad B. Normalized Testosterone Glucuronide as a Potential Urinary Biomarker for Highly Variable UGT2B17 in Children 7-18 Years. Clin Pharmacol Ther 2020; 107:1149-1158. [PMID: 31900930 DOI: 10.1002/cpt.1764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022]
Abstract
UDP-glucuronosyltransferase 2B17 (UGT2B17) is a highly variable androgen-metabolizing and drug-metabolizing enzyme. UGT2B17 exhibits a unique ontogeny profile characterized by a dramatic increase in hepatic protein expression from prepubertal age to adulthood. Age, sex, copy number variation (CNV), and single nucleotide polymorphisms only explain 26% of variability in protein expression, highlighting the need for a phenotypic biomarker for predicting interindividual variability in glucuronidation of UGT2B17 substrates. Here, we propose testosterone glucuronide (TG) normalized by androsterone glucuronide (TG/AG) as a urinary UGT2B17 biomarker, and examine the associations among urinary TG/AG and age, sex, and CNV. We performed targeted metabolomics of 12 androgen conjugates with liquid-chromatography tandem mass spectrometry in 63 pediatric subjects ages 7-18 years followed over 7 visits in 3 years. Consistent with the reported developmental trajectory of UGT2B17 protein expression, urinary TG/AG is significantly associated with age, sex, and CNV. In conclusion, TG/AG shows promise as a phenotypic urinary UGT2B17 biomarker.
Collapse
Affiliation(s)
- Haeyoung Zhang
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Abdul Basit
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Chris Wolford
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Kuan-Fu Chen
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Yvonne S Lin
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
14
|
Wang YQ, Shang XF, Wang L, Zhang P, Zou LW, Song YQ, Hao DC, Fang SQ, Ge GB, Tang H. Interspecies variation of clopidogrel hydrolysis in liver microsomes from various mammals. Chem Biol Interact 2019; 315:108871. [PMID: 31669218 DOI: 10.1016/j.cbi.2019.108871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/09/2019] [Accepted: 10/21/2019] [Indexed: 02/08/2023]
Abstract
Clopidogrel, a clinically used antiplatelet agent, can be readily hydrolyzed by human carboxylesterase 1A (CES1A) to release an inactive metabolite clopidogrel carboxylic acid (CCA). In this study, clopidogrel was used as a tool substrate to investigate the interspecies variation of clopidogrel hydrolysis in hepatic microsomes from various mammals including human and six laboratory animals (such as mouse, rat, rabbit, beagle dog, minipig and cynomolgus monkey). The results demonstrated that clopidogrel could be hydrolyzed into CCA by all tested hepatic microsomes from human or other mammals, but the hydrolytic rates greatly varied among species. Inhibition assays demonstrated that BNPP (an inactivator of mammalian CES) strongly inactivated clopidogrel hydrolytic activity in all tested hepatic microsomes, suggested that mammalian CES were major contributor(s) responsible for clopidogrel hydrolysis in hepatic preparations from all above-mentioned species. By contrast, the response of a reversible inhibitor of human CES1A on clopidogrel hydrolysis in these liver preparations varied significantly among different species. Moreover, the enzymatic kinetics and the apparent kinetic parameters of clopidogrel hydrolysis in hepatic microsomes from various animal species were evaluated and compared to each other. These findings provide crucial information for deeply understanding the differences in catalytic behaviors of mammalian CES, which will be very helpful for choosing suitable laboratory animal(s) for whole tests of CES1A substrate-drugs.
Collapse
Affiliation(s)
- Ya-Qiao Wang
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, 832000, China
| | - Xiao-Feng Shang
- Zhangye People's Hospital affiliated to Hexi University, Zhangye, Gansu, 734000, China
| | - Lu Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, 832000, China
| | - Ping Zhang
- Department of Cardiology, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Li-Wei Zou
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China
| | - Yun-Qing Song
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China
| | - Da-Cheng Hao
- Dalian Jiaotong University, Dalian, 116028, China
| | - Sheng-Quan Fang
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China
| | - Guang-Bo Ge
- Translational Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine & Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China.
| | - Hui Tang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Pharmacy School of Shihezi University, Xinjiang, 832000, China.
| |
Collapse
|
15
|
Deguchi S, Yamashita T, Igai K, Harada K, Toba Y, Hirata K, Takayama K, Mizuguchi H. Modeling of Hepatic Drug Metabolism and Responses in CYP2C19 Poor Metabolizer Using Genetically Manipulated Human iPS cells. Drug Metab Dispos 2019; 47:632-638. [PMID: 30962288 DOI: 10.1124/dmd.119.086322] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/02/2019] [Indexed: 08/30/2023] Open
Abstract
Cytochrome P450 family 2 subfamily C member 19 (CYP2C19), in liver, plays important roles in terms of drug metabolism. It is known that CYP2C19 poor metabolizers (PMs) lack CYP2C19 metabolic capacity. Thus, unexpected drug-induced liver injury or decrease of drug efficacy would be caused in CYP2C19 substrate-treated CYP2C19 PMs. However, it is difficult to evaluate the safety and effectiveness of drugs and candidate compounds for CYP2C19 PMs because there is currently no model for this phenotype. Here, using human induced pluripotent stem cells (human iPS cells) and our highly efficient genome-editing and hepatocyte differentiation technologies, we generated CYP2C19-knockout human iPS cell-derived hepatocyte-like cells (CYP2C19-KO HLCs) as a novel CYP2C19 PM model for drug development and research. The gene expression levels of hepatocyte markers were similar between wild-type iPS cell-derived hepatocyte-like cells (WT HLCs) and CYP2C19-KO HLCs, suggesting that CYP2C19 deficiency did not affect the hepatic differentiation potency. We also examined CYP2C19 metabolic activity by measuring S-mephenytoin metabolites using ultra-performance liquid chromatography-tandem mass spectrometry. The CYP2C19 metabolic activity was almost eliminated by CYP2C19 knockout. Additionally, we evaluated whether clopidogrel (CYP2C19 substrate)-induced liver toxicity could be predicted using our model. Unexpectedly, there was no significant difference in cell viability between clopidogrel-treated WT HLCs and CYP2C19-KO HLCs. However, the cell viability in clopidogrel- and ketoconazole (CYP3A4 inhibitor)-treated CYP2C19-KO HLCs was significantly enhanced as compared with that in clopidogrel- and DMSO-treated CYP2C19-KO HLCs. This result suggests that CYP2C19-KO HLCs can predict clopidogrel-induced liver toxicity. We succeeded in generating CYP2C19 PM model cells using human iPS cells and genome-editing technologies for pharmaceutical research. SIGNIFICANCE STATEMENT: Although unexpected drug-induced liver injury or decrease of drug efficacy would be caused in CYP2C19 substrate-treated CYP2C19 poor metabolizers, it is difficult to evaluate the safety and effectiveness of drugs and candidate compounds for CYP2C19 poor metabolizers because there is currently no model for this phenotype. Using human iPS cells and our highly efficient genome editing and hepatocyte differentiation technologies, we generated CYP2C19-knockout human iPS cell-derived hepatocyte-like cells as a novel CYP2C19 poor metabolizer model for drug development and research.
Collapse
Affiliation(s)
- Sayaka Deguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| | - Tomoki Yamashita
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| | - Keisuke Igai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| | - Kazuo Harada
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| | - Yukiko Toba
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| | - Kazumasa Hirata
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| | - Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences (S.D., T.Y., K.I., Y.T., K.T., H.M.), Laboratory of Applied Environmental Biology, Graduate School of Pharmaceutical Sciences (K.Ha., K.Hi.), and Global Center for Medical Engineering and Informatics (H.M.), Osaka University, Osaka, Japan; PRESTO, Japan Science and Technology Agency, Saitama, Japan (K.T.); and Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan (Y.T., K.T., H.M.)
| |
Collapse
|
16
|
Émond JP, Labriet A, Desjardins S, Rouleau M, Villeneuve L, Hovington H, Brisson H, Lacombe L, Simonyan D, Caron P, Périgny M, Têtu B, Fallon JK, Klein K, Smith PC, Zanger UM, Guillemette C, Lévesque E. Factors Affecting Interindividual Variability of Hepatic UGT2B17 Protein Expression Examined Using a Novel Specific Monoclonal Antibody. Drug Metab Dispos 2019; 47:444-452. [PMID: 30819787 DOI: 10.1124/dmd.119.086330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 02/21/2019] [Indexed: 08/10/2024] Open
Abstract
Accurate quantification of the metabolic enzyme uridine diphospho-glucuronosyltransferase (UGT) UGT2B17 has been hampered by the high sequence identity with other UGT2B enzymes (as high as 94%) and by the lack of a specific antibody. Knowing the significance of the UGT2B17 pathway in drug and hormone metabolism and cancer, we developed a specific monoclonal antibody (EL-2B17mAb), initially validated by the lack of detection in liver microsomes of an individual carrying no UGT2B17 gene copy and in supersomes expressing UGT2B enzymes. Immunohistochemical detection in livers revealed strong labeling of bile ducts and variable labeling of hepatocytes. Expression levels assessed by immunoblotting were highly correlated to mass spectrometry-based quantification (r = 0.93), and three major expression patterns (absent, low, or high) were evidenced. Livers with very low expression were carriers of the functional rs59678213 G variant, located in the binding site for the transcription factor forkhead box A1 (FOXA1) of the UGT2B17 promoter. The highest level of expression was observed for individuals carrying at least one rs59678213 A allele. Multiple regression analysis indicated that the number of gene copies explained only 8% of UGT2B17 protein expression, 49% when adding rs59678213, reaching 54% when including sex. The novel EL-2B17mAb antibody allowed specific UGT2B17 quantification and exposed different patterns of hepatic expression. It further suggests that FOXA1 is a key driver of UGT2B17 expression in the liver. The availability of this molecular tool will help characterize the UGT2B17 level in various disease states and establish more precisely the contribution of the UGT2B17 enzyme to drug and hormone metabolism.
Collapse
Affiliation(s)
- Jean-Philippe Émond
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Adrien Labriet
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Sylvie Desjardins
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Michèle Rouleau
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Lyne Villeneuve
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Hélène Hovington
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Hervé Brisson
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Louis Lacombe
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - David Simonyan
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Patrick Caron
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Martine Périgny
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Bernard Têtu
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - John K Fallon
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Kathrin Klein
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Philip C Smith
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Ulrich M Zanger
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Chantal Guillemette
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| | - Eric Lévesque
- Centre Hospitalier Universitaire (CHU) de Québec Research Centre and Faculty of Medicine (J.-P.É., S.D., H.H., H.B., L.L., M.P., B.T., E.L.) and CHU de Québec Research Centre and Faculty of Pharmacy, Laval University (A.L., M.R., L.V., P.C., C.G.), and Statistical and Clinical Research Platform, CHU de Québec Research Centre (D.S.), Québec, Canada.); Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S.); and Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany (K.K., U.M.Z.)
| |
Collapse
|
17
|
Kahma H, Filppula AM, Launiainen T, Viinamäki J, Neuvonen M, Evangelista EA, Totah RA, Backman JT. Critical Differences between Enzyme Sources in Sensitivity to Detect Time-Dependent Inactivation of CYP2C8. Drug Metab Dispos 2019; 47:436-443. [PMID: 30709838 PMCID: PMC11022894 DOI: 10.1124/dmd.118.085498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Clopidogrel acyl-β-d-glucuronide is a mechanism-based inhibitor of cytochrome P450 2C8 in human liver microsomes (HLMs). However, time-dependent inactivation (TDI) of CYP2C8 could not be detected in an earlier study in human recombinant CYP2C8 (Supersomes). Here, we investigate whether different enzyme sources exhibit differences in detection of CYP2C8 TDI under identical experimental conditions. Inactivation of CYP2C8 by amiodarone (100 μM), clopidogrel acyl-β-d-glucuronide (100 μM), gemfibrozil 1-O-β-glucuronide (100 μM), and phenelzine (100 μM) was investigated in HLMs and three recombinant human CYP2C8 preparations (Supersomes, Bactosomes, and EasyCYP Bactosomes) using amodiaquine N-deethylation as the marker reaction. Furthermore, the inactivation kinetics of CYP2C8 by clopidogrel glucuronide (5-250 μM) was determined in Supersomes and Bactosomes. Amiodarone caused weak TDI in all enzyme preparations tested, while the extent of inactivation by clopidogrel glucuronide, gemfibrozil glucuronide, and phenelzine varied markedly between preparations, and even different Supersome lots. Both glucuronides caused strong inactivation of CYP2C8 in HLMs, Bactosomes and in one Supersome lot (>50% inhibition), but significant inactivation could not be reliably detected in other Supersome lots or EasyCYP Bactosomes. In Bactosomes, the concentration producing half of kinact (KI) and maximal inactivation rate (kinact) of clopidogrel glucuronide (14 μM and 0.054 minute-1) were similar to those determined previously in HLMs. Phenelzine caused strong inactivation of CYP2C8 in one Supersome lot (91% inhibition) but not in HLMs or other recombinant CYP2C8 preparations. In conclusion, different enzyme sources and different lots of the same recombinant enzyme preparation are not equally sensitive to detect inactivation of CYP2C8, suggesting that recombinant CYPs should be avoided when identifying mechanism-based inhibitors.
Collapse
Affiliation(s)
- Helinä Kahma
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Anne M Filppula
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Terhi Launiainen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Jenni Viinamäki
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Mikko Neuvonen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Eric A Evangelista
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Rheem A Totah
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Janne T Backman
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| |
Collapse
|
18
|
Zhang H, Basit A, Busch D, Yabut K, Bhatt DK, Drozdzik M, Ostrowski M, Li A, Collins C, Oswald S, Prasad B. Quantitative characterization of UDP-glucuronosyltransferase 2B17 in human liver and intestine and its role in testosterone first-pass metabolism. Biochem Pharmacol 2018; 156:32-42. [PMID: 30086285 PMCID: PMC6188809 DOI: 10.1016/j.bcp.2018.08.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022]
Abstract
Protein abundance and activity of UGT2B17, a highly variable drug- and androgen-metabolizing enzyme, were quantified in microsomes, S9 fractions, and primary cells isolated from human liver and intestine by validated LC-MS/MS methods. UGT2B17 protein abundance showed >160-fold variation (mean ± SD, 1.7 ± 2.7 pmol/mg microsomal protein) in adult human liver microsomes (n = 26) and significant correlation (r2 = 0.77, p < 0.001) with testosterone glucuronide (TG) formation. Primary role of UGT2B17 in TG formation compared to UGT2B15 was confirmed by performing activity assays in UGT2B17 gene deletion samples and with a selective UGT2B17 inhibitor, imatinib. Human intestinal microsomes isolated from small intestine (n = 6) showed on average significantly higher protein abundance (7.4 ± 6.6 pmol/mg microsomal protein, p = 0.016) compared to liver microsomes, with an increasing trend towards distal segments of the gastrointestinal (GI) tract. Commercially available pooled microsomes and S9 fractions confirmed greater abundance and activity of UGT2B17 in intestinal fractions compared to liver fractions. To further investigate the quantitative role of UGT2B17 in testosterone metabolism in whole cell system, a targeted metabolomics study was performed in hepatocytes (n = 5) and enterocytes (n = 16). TG was the second most abundant metabolite after androstenedione in both cell systems. Reasonable correlation between UGT2B17 abundance and activity were observed in enterocytes (r2 = 0.69, p = 0.003), but not in hepatocytes. These observational and mechanistic data will be useful in developing physiologically-based pharmacokinetic (PBPK) models for predicting highly-variable first-pass metabolism of testosterone and other UGT2B17 substrates.
Collapse
Affiliation(s)
- Haeyoung Zhang
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Abdul Basit
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Diana Busch
- Department of Clinical Pharmacology, University of Greifswald, Greifswald, Germany
| | - King Yabut
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | | | - Marek Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Marek Ostrowski
- Department of General and Transplantation Surgery, Pomeranian Medical University, Szczecin, Poland
| | - Albert Li
- In Vitro ADMET Laboratories (IVAL), Columbia, MD, USA
| | - Carol Collins
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Stefan Oswald
- Department of Clinical Pharmacology, University of Greifswald, Greifswald, Germany
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
19
|
Basit A, Amory JK, Prasad B. Effect of Dose and 5α-Reductase Inhibition on the Circulating Testosterone Metabolite Profile of Men Administered Oral Testosterone. Clin Transl Sci 2018; 11:513-522. [PMID: 29877607 PMCID: PMC6132360 DOI: 10.1111/cts.12569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/11/2018] [Indexed: 12/16/2022] Open
Abstract
Development of an oral testosterone therapy has proven extremely challenging because of extensive and variable first-pass metabolism. We investigated the in vivo metabolism of testosterone with increasing oral doses of testosterone, both alone and with the co-administration of dutasteride (5α-reductase inhibitor) by liquid-chromatography tandem mass spectrometry (LC-MS/MS). In eugonadal men prior to dosing, the circulating concentration of testosterone, androstenedione, etiocholanolone-glucuronide, and androsterone-glucuronide was 8.6, 20.9, 9.1, and 55.3%, respectively, of the total testosterone-related species, whereas testosterone-glucuronide was ∼1%. When testosterone was dosed orally to men with experimental hypogonadism, a proportion of testosterone-glucuronide increased to 13%. Dutasteride treatment significantly decreased levels of androsterone and its metabolites. This work reveals extensive metabolism of orally dosed testosterone to androsterone glucuronide via androstenedione, with testosterone-glucuronide appearing to be the second most important metabolite. This information is of importance in the development of an effective oral testosterone therapy and may have implications for testosterone doping research.
Collapse
Affiliation(s)
- Abdul Basit
- Department of PharmaceuticsUniversity of WashingtonSeattle, WashingtonUSA
| | - John K. Amory
- Department of MedicineUniversity of WashingtonSeattle, WashingtonUSA
| | - Bhagwat Prasad
- Department of PharmaceuticsUniversity of WashingtonSeattle, WashingtonUSA
| |
Collapse
|
20
|
Holmberg MT, Tornio A, Paile-Hyvärinen M, Tarkiainen EK, Neuvonen M, Neuvonen PJ, Backman JT, Niemi M. CYP3A4*22 Impairs the Elimination of Ticagrelor, But Has No Significant Effect on the Bioactivation of Clopidogrel or Prasugrel. Clin Pharmacol Ther 2018; 105:448-457. [PMID: 29998574 DOI: 10.1002/cpt.1177] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/03/2018] [Indexed: 11/09/2022]
Abstract
CYP3A enzymes participate in the elimination of ticagrelor and the bioactivation of clopidogrel and prasugrel. We studied the effects of functional CYP3A genetic variants (CYP3A4*22; rs35599367 and CYP3A5*3; rs776746) on the pharmacokinetics and pharmacodynamics of ticagrelor, clopidogrel, and prasugrel. Six healthy volunteers with the CYP3A4*1/*22 and CYP3A5*3/*3 genotype (CYP3A4*22 carriers), eight with the CYP3A4*1/*1 and CYP3A5*1/*3 genotype (CYP3A5 expressors), and 11-13 with the CYP3A4*1/*1 and CYP3A5*3/*3 genotypes (controls) ingested single doses of ticagrelor, clopidogrel, and prasugrel on separate occasions. Ticagrelor area under the plasma concentration-time curve (AUC) was 89% (P = 0.004) higher in CYP3A4*22 carriers than in controls. CYP3A4*22 carriers also showed more pronounced platelet inhibition at 24 hours after ticagrelor ingestion than the controls (43% vs. 21%; P = 0.029). The CYP3A5 genotype did not affect ticagrelor pharmacokinetics. Neither CYP3A5 nor CYP3A4 genotypes significantly affected prasugrel or clopidogrel. In conclusion, the CYP3A4*22 allele markedly impairs ticagrelor elimination enhancing its antiplatelet effect.
Collapse
Affiliation(s)
- Mikko T Holmberg
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Maria Paile-Hyvärinen
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - E Katriina Tarkiainen
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
21
|
Itkonen MK, Tornio A, Lapatto-Reiniluoto O, Neuvonen M, Neuvonen PJ, Niemi M, Backman JT. Clopidogrel Increases Dasabuvir Exposure With or Without Ritonavir, and Ritonavir Inhibits the Bioactivation of Clopidogrel. Clin Pharmacol Ther 2018; 105:219-228. [PMID: 29696643 PMCID: PMC6585621 DOI: 10.1002/cpt.1099] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/23/2018] [Indexed: 12/11/2022]
Abstract
Dasabuvir is mainly metabolized by cytochrome P450 (CYP) 2C8 and is predominantly used in a regimen containing ritonavir. Ritonavir and clopidogrel are inhibitors of CYP3A4 and CYP2C8, respectively. In a randomized, crossover study in 12 healthy subjects, we examined the impact of clinical doses of ritonavir (for 5 days), clopidogrel (for 3 days), and their combination on dasabuvir pharmacokinetics, and the effect of ritonavir on clopidogrel. Clopidogrel, but not ritonavir, increased the geometric mean AUC0‐∞ of dasabuvir 4.7‐fold; range 2.0–10.1‐fold (P = 8·10−7), compared with placebo. Clopidogrel and ritonavir combination increased dasabuvir AUC0‐∞ 3.9‐fold; range 2.1–7.9‐fold (P = 2·10−6), compared with ritonavir alone. Ritonavir decreased the AUC0‐4h of clopidogrel active metabolite by 51% (P = 0.0001), and average platelet inhibition from 51% without ritonavir to 31% with ritonavir (P = 0.0007). In conclusion, clopidogrel markedly elevates dasabuvir concentrations, and patients receiving ritonavir are at risk for diminished clopidogrel response.
Collapse
Affiliation(s)
- Matti K Itkonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Outi Lapatto-Reiniluoto
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|