1
|
Jiang Z, Wang Y, Zhao G, Luo X, Shen Y, Li W, Peng Y, Zheng J. Metabolic Activation of Stiripentol Correlates with Cytotoxicity. Chem Res Toxicol 2025. [PMID: 39924957 DOI: 10.1021/acs.chemrestox.4c00209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Stiripentol (SRP) is an antiepileptic agent utilized in managing seizures related to Dravet syndrome. Long-term safety studies have highlighted significant adverse effects in patients including drowsiness, reduced appetite, ataxia, and elevated levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST). The present study aimed at identifying the reactive metabolite of SRP and defining the potential correlation between its cytotoxicity and metabolic activation. Rat liver microsome incubation of SRP fortified with GSH as a trapping agent produced an α,β-unsaturated ketone metabolite (M1) and a related GSH conjugate (M2). Moreover, both the phase I metabolite and the GSH conjugate were detected in the bile of SRP-treated rats, indicating that both in vivo and in vitro metabolic activation of SRP took place. Notably, SRP exhibited significant cytotoxicity toward rat primary hepatocytes. Pretreatment with ketoconazole, a selective CYP3A enzyme inhibitor, mitigated the susceptibility of hepatocytes to SRP-induced cytotoxicity. These findings suggest that SRP may undergo metabolism to the α,β-unsaturated ketone metabolite, potentially contributing to the cytotoxic effects associated with SRP.
Collapse
Affiliation(s)
- Ziying Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Yang Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Guode Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Xinyu Luo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Yan Shen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Weiwei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China
| |
Collapse
|
2
|
Guengerich FP. Roles of Individual Human Cytochrome P450 Enzymes in Drug Metabolism. Pharmacol Rev 2024; 76:1104-1132. [PMID: 39054072 PMCID: PMC11549934 DOI: 10.1124/pharmrev.124.001173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/28/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Our knowledge of the roles of individual cytochrome P450 (P450) enzymes in drug metabolism has developed considerably in the past 30 years, and this base has been of considerable use in avoiding serious issues with drug interactions and issues due to variations. Some newer approaches are being considered for "phenotyping" metabolism reactions with new drug candidates. Endogenous biomarkers are being used for noninvasive estimation of levels of individual P450 enzymes. There is also the matter of some remaining "orphan" P450s, which have yet to be assigned reactions. Practical problems that continue in drug development include predicting drug-drug interactions, predicting the effects of polymorphic and other P450 variations, and evaluating interspecies differences in drug metabolism, particularly in the context of "metabolism in safety testing" regulatory issues ["disproportionate (human) metabolites"]. SIGNIFICANCE STATEMENT: Cytochrome P450 enzymes are the major catalysts involved in drug metabolism. The characterization of their individual roles has major implications in drug development and clinical practice.
Collapse
Affiliation(s)
- F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
3
|
Bauman JN, Doran AC, Gualtieri GM, Hee B, Strelevitz T, Cerny MA, Banfield C, Plotka A, Wang X, Purohit VS, Dowty ME. The Pharmacokinetics, Metabolism, and Clearance Mechanisms of Ritlecitinib, a Janus Kinase 3 and Tyrosine-Protein Kinase Family Inhibitor, in Humans. Drug Metab Dispos 2024; 52:1124-1136. [PMID: 39111823 DOI: 10.1124/dmd.124.001843] [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: 06/13/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024] Open
Abstract
Ritlecitinib is an oral once-daily irreversible inhibitor of Janus kinase 3 and tyrosine-protein kinase family being developed for the treatment of moderate-to-severe alopecia areata. This study examined the disposition of ritlecitinib in male participants following oral and intravenous administration using accelerator mass spectroscopy methodology to estimate pharmacokinetic parameters and characterize metabolite profiles. The results indicated ritlecitinib had a systemic clearance of 43.7 L/h, a steady state volume of distribution of 73.8 L, extent of absorption of 89%, time to maximum plasma concentration of ∼0.5 hours, and absolute oral bioavailability of 64%. An observed long terminal half-life of total radioactivity was primarily attributed to ritlecitinib binding to plasma albumin. Ritlecitinib was the main circulating drug species in plasma (∼30%), with one major pharmacologically inactive cysteine conjugated metabolite (M2) at >10%. Oxidative metabolism (fractional clearance 0.47) and glutathione-related conjugation (fractional clearance 0.24) were the primary routes of elimination for ritlecitinib with the greatest disposition of radioactivity shown in the urine (∼71%). In vitro phenotyping indicated ritlecitinib cytochrome P450 (CYP) fraction of metabolism assignments of 0.29 for CYP3A, 0.09 for CYP2C8, 0.07 for CYP1A2, and 0.02 for CYP2C9. In vitro phenotyping in recombinant human glutathione S-transferases indicated ritlecitinib was turned over by a number of cytosolic and microsomal enzyme isoforms. SIGNIFICANCE STATEMENT: This study provides a detailed understanding of the disposition and metabolism of ritlecitinib, a JAK3 and TEC family kinase inhibitor for alopecia areata in humans, as well as characterization of clearance pathways and pharmacokinetics of ritlecitinib and its metabolites. As an AMS-based ADME study design, we have expanded on reporting the standard ADME endpoints, providing key pharmacokinetic parameters, such as clearance, volume of distribution, and bioavailability, allowing for a more comprehensive understanding of drug disposition.
Collapse
Affiliation(s)
- Jonathan N Bauman
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Angela C Doran
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Gabrielle M Gualtieri
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Brian Hee
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Timothy Strelevitz
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Matthew A Cerny
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Christopher Banfield
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Anna Plotka
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Xiaoxing Wang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Vivek S Purohit
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| | - Martin E Dowty
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Cambridge, Massachusetts (M.E.D.) andGroton, Connecticut (J.N.B., A.C.D., G.M.G., B.H., T.S., M.A.C.); Clinical Pharmacology, Pfizer, Inc., Groton, Connecticut (C.B., X.W., V.S.P.); Biostatistics, Pfizer, Inc., Collegeville, Pennsylvania (A.P.)
| |
Collapse
|
4
|
Lapham K, Ferguson N, Niosi M, Goosen TC. Clotrimazole Identified as a Selective UGT2B4 Inhibitor Using Canagliflozin-2'- O-Glucuronide Formation as a Selective UGT2B4 Probe Reaction. Drug Metab Dispos 2024; 52:1083-1093. [PMID: 39142826 DOI: 10.1124/dmd.124.001812] [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: 05/20/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
UGT2B4 is a highly expressed drug-metabolizing enzyme in the liver contributing to the glucuronidation of several drugs. To enable quantitatively assessing UGT2B4 contribution toward metabolic clearance, a potent and selective UGT2B4 inhibitor that can be used for reaction phenotyping was sought. Initially, a canagliflozin-2'-O-glucuronyl transferase activity assay was developed in recombinant UGT2B4 and human liver microsomes (HLM) [±2% bovine serum albumin (BSA)]. Canagliflozin-2'-O-glucuronidation (C2OG) substrate concentration at half-maximal velocity value in recombinant UGT2B4 and HLM were similar. C2OG formation intrinsic clearance was five- to seven-fold higher in incubations containing 2% BSA, suggesting UGT2B4 susceptibility to the inhibitory unsaturated long-chain fatty acids released during the incubation. Monitoring for C2OG formation, 180 compounds were evaluated for UGT2B4 inhibition potency in the presence and absence of 2% BSA. Compounds that exhibited an apparent UGT2B4 IC50 of < 1 μM in HLM with 2% BSA were evaluated for inhibition of UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2B7, UGT2B10, UGT2B15, and UGT2B17 catalytic activities to establish selectivity suitable for supporting UGT reaction phenotyping. In this study, clotrimazole was identified as a potent UGT2B4 inhibitor (HLM apparent IC50 of 11 to 35 nM ± 2% BSA). Moreover, clotrimazole exhibited selectivity for UGT2B4 inhibition (>24-fold) over the other UGT enzymes evaluated. Additionally, during this study it was discovered that the previously described UGT2B7 inhibitors 16α- and 16β-phenyllongifolol also inhibit UGT2B4. Clotrimazole, a potent and selective UGT2B4 inhibitor, will prove essential during UGT reaction phenotyping. SIGNIFICANCE STATEMENT: To mechanistically evaluate drug interactions, it is essential to understand the contribution of individual enzymes to the metabolic clearance of a drug. The present study describes the development of a UGT2B4 activity assay that enabled the discovery of the highly selective and potent UGT2B4 inhibitor clotrimazole. Clotrimazole can be used in UGT reaction phenotyping studies to estimate fractional contribution of UGT2B4.
Collapse
Affiliation(s)
- Kimberly Lapham
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| | - Nicholas Ferguson
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| | - Mark Niosi
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| | - Theunis C Goosen
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Groton, Connecticut
| |
Collapse
|
5
|
Garnsey MR, Robinson MC, Nguyen LT, Cardin R, Tillotson J, Mashalidis E, Yu A, Aschenbrenner L, Balesano A, Behzadi A, Boras B, Chang JS, Eng H, Ephron A, Foley T, Ford KK, Frick JM, Gibson S, Hao L, Hurst B, Kalgutkar AS, Korczynska M, Lengyel-Zhand Z, Gao L, Meredith HR, Patel NC, Polivkova J, Rai D, Rose CR, Rothan H, Sakata SK, Vargo TR, Qi W, Wu H, Liu Y, Yurgelonis I, Zhang J, Zhu Y, Zhang L, Lee AA. Discovery of SARS-CoV-2 papain-like protease (PL pro) inhibitors with efficacy in a murine infection model. SCIENCE ADVANCES 2024; 10:eado4288. [PMID: 39213347 PMCID: PMC11364104 DOI: 10.1126/sciadv.ado4288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Vaccines and first-generation antiviral therapeutics have provided important protection against COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, there remains a need for additional therapeutic options that provide enhanced efficacy and protection against potential viral resistance. The SARS-CoV-2 papain-like protease (PLpro) is one of the two essential cysteine proteases involved in viral replication. While inhibitors of the SARS-CoV-2 main protease have demonstrated clinical efficacy, known PLpro inhibitors have, to date, lacked the inhibitory potency and requisite pharmacokinetics to demonstrate that targeting PLpro translates to in vivo efficacy in a preclinical setting. Here, we report the machine learning-driven discovery of potent, selective, and orally available SARS-CoV-2 PLpro inhibitors, with lead compound PF-07957472 (4) providing robust efficacy in a mouse-adapted model of COVID-19 infection.
Collapse
Affiliation(s)
| | | | | | - Rhonda Cardin
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Joseph Tillotson
- Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | | | - Aijia Yu
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | | | - Amanda Balesano
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Amin Behzadi
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Britton Boras
- Pfizer Global Research and Development, La Jolla, CA 92121, USA
| | - Jeanne S. Chang
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Heather Eng
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Andrew Ephron
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Tim Foley
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Kristen K. Ford
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - James M. Frick
- PostEra, 1 Broadway, 14th floor, Cambridge, MA 02142, USA
| | - Scott Gibson
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences,Utah State University, Logan, UT 84322, USA
| | - Li Hao
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Brett Hurst
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences,Utah State University, Logan, UT 84322, USA
| | | | | | | | - Liping Gao
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | | | - Nandini C. Patel
- Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Jana Polivkova
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Devendra Rai
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Colin R. Rose
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Hussin Rothan
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | | | | | - Wenying Qi
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | - Huixian Wu
- Pfizer Global Research and Development, Groton, CT 06340, USA
| | - Yiping Liu
- WuXi, WuXi AppTec (Shanghai) Co. Ltd. Shanghai 200131, China
| | - Irina Yurgelonis
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Jinzhi Zhang
- Pfizer Global Research and Development, Shanghai 201210, China
| | - Yuao Zhu
- Pfizer Global Research and Development, Pearl River, NY 10965, USA
| | - Lei Zhang
- Pfizer Global Research and Development, Cambridge, MA 02139, USA
| | - Alpha A. Lee
- PostEra, 1 Broadway, 14th floor, Cambridge, MA 02142, USA
| |
Collapse
|
6
|
Luo LJ, Liu X, Li Y, Li Y, Sheng L. Characterization of the metabolic contributions of cytochrome P450 isoforms to bicyclol using the relative activity factor method. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:918-929. [PMID: 38629733 DOI: 10.1080/10286020.2024.2340072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/02/2024] [Indexed: 06/27/2024]
Abstract
Bicyclol is a hepatoprotective agent widely used for treating chronic hepatitis and drug-induced liver injuries in clinics. The purpose of the study was to elucidate the contribution of CYP450 enzymes to the metabolism of bicyclol using the relative activity factor approach. After incubation with human liver microsomes and recombinant human liver CYP450 enzymes, the calculated contribution of CYP3A4 and 2C19 to the metabolism of bicyclol was 85.6-90.3% and 9.2-9.7%, respectively. The metabolism was interrupted in the presence of CYP3A4 and 2C19 selective inhibitors. These findings help to predict or avoid metabolic drug-drug interactions or toxicity in clinical applications of bicyclol.
Collapse
Affiliation(s)
- Li-Jun Luo
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiao Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yang Li
- Beijing Union Pharmaceutical Factory, Beijing 102600, China
| | - Li Sheng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Drug Metabolism, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
7
|
Yadav J, Maldonato BJ, Roesner JM, Vergara AG, Paragas EM, Aliwarga T, Humphreys S. Enzyme-mediated drug-drug interactions: a review of in vivo and in vitro methodologies, regulatory guidance, and translation to the clinic. Drug Metab Rev 2024:1-33. [PMID: 39057923 DOI: 10.1080/03602532.2024.2381021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Enzyme-mediated pharmacokinetic drug-drug interactions can be caused by altered activity of drug metabolizing enzymes in the presence of a perpetrator drug, mostly via inhibition or induction. We identified a gap in the literature for a state-of-the art detailed overview assessing this type of DDI risk in the context of drug development. This manuscript discusses in vitro and in vivo methodologies employed during the drug discovery and development process to predict clinical enzyme-mediated DDIs, including the determination of clearance pathways, metabolic enzyme contribution, and the mechanisms and kinetics of enzyme inhibition and induction. We discuss regulatory guidance and highlight the utility of in silico physiologically-based pharmacokinetic modeling, an approach that continues to gain application and traction in support of regulatory filings. Looking to the future, we consider DDI risk assessment for targeted protein degraders, an emerging small molecule modality, which does not have recommended guidelines for DDI evaluation. Our goal in writing this report was to provide early-career researchers with a comprehensive view of the enzyme-mediated pharmacokinetic DDI landscape to aid their drug development efforts.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism & Bioanalytics (PDMB), Merck & Co., Inc., Boston, MA, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Joseph M Roesner
- Department of Pharmacokinetics, Dynamics, Metabolism & Bioanalytics (PDMB), Merck & Co., Inc., Boston, MA, USA
| | - Ana G Vergara
- Department of Pharmacokinetics, Dynamics, Metabolism & Bioanalytics (PDMB), Merck & Co., Inc., Rahway, NJ, USA
| | - Erickson M Paragas
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| | - Theresa Aliwarga
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| | - Sara Humphreys
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, South San Francisco, CA, USA
| |
Collapse
|
8
|
Kukla DA, Belair DG, Stresser DM. Evaluation and Optimization of a Microcavity Plate-Based Human Hepatocyte Spheroid Model for Predicting Clearance of Slowly Metabolized Drug Candidates. Drug Metab Dispos 2024; 52:797-812. [PMID: 38777596 DOI: 10.1124/dmd.124.001653] [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: 01/23/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
In vitro clearance assays are routinely conducted in drug discovery to predict in vivo clearance, but low metabolic turnover compounds are often difficult to evaluate. Hepatocyte spheroids can be cultured for days, achieving higher drug turnover, but have been hindered by limitations on cell number per well. Corning Elplasia microcavity 96-well microplates enable the culture of 79 hepatocyte spheroids per well. In this study, microcavity spheroid properties (size, hepatocyte function, longevity, culturing techniques) were assessed and optimized for clearance assays, which were then compared with microsomes, hepatocyte suspensions, two-dimensional-plated hepatocytes, and macrowell spheroids cultured as one per well. Higher enzyme activity coupled with greater hepatocyte concentrations in microcavity spheroids enabled measurable turnover of all 17 test compounds, unlike the other models that exhibited less drug turnover. Microcavity spheroids also predicted intrinsic clearance (CLint) and blood clearance (CLb) within threefold for 53% [9/17; average absolute fold error (AAFE), 3.9] and 82% (14/17; AAFE, 2.6) of compounds using a linear regression correction model, respectively. An alternate method incorporating mechanistic modeling that accounts for mass transport (permeability and diffusion) within spheroids demonstrated improved predictivity for CLint (12/17; AAFE, 4.0) and CLb (14/17; AAFE, 2.1) without the need for empirical scaling factors. The estimated fraction of drug metabolized by cytochrome P450 3A4 (fm,CYP3A4) using 3 μM itraconazole was within 25% of observed values for 6 of 8 compounds, with 5 of 8 compounds within 10%. In sum, spheroid cultures in microcavity plates permit the ability to test and predict clearance as well as fm,CYP3A4 of low metabolic turnover compounds and represent a valuable complement to conventional in vitro clearance assays. SIGNIFICANCE STATEMENT: Culturing multiple spheroids in ultralow attachment microcavities permits accurate quantitation of metabolically stable compounds in substrate depletion assays, overcoming limitations with singly cultured spheroids. In turn, this permits robust estimates of intrinsic clearance, which is improved with the consideration of mass transport within the spheroid. Incubations with 3 μM itraconazole enabled assessments of CYP3A4 involvement in hepatic clearance.
Collapse
Affiliation(s)
- David A Kukla
- Quantitative, Translational, and ADME Sciences, AbbVie Inc., North Chicago, Illinois
| | - David G Belair
- Quantitative, Translational, and ADME Sciences, AbbVie Inc., North Chicago, Illinois
| | - David M Stresser
- Quantitative, Translational, and ADME Sciences, AbbVie Inc., North Chicago, Illinois
| |
Collapse
|
9
|
Lee J, Beers JL, Geffert RM, Jackson KD. A Review of CYP-Mediated Drug Interactions: Mechanisms and In Vitro Drug-Drug Interaction Assessment. Biomolecules 2024; 14:99. [PMID: 38254699 PMCID: PMC10813492 DOI: 10.3390/biom14010099] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Drug metabolism is a major determinant of drug concentrations in the body. Drug-drug interactions (DDIs) caused by the co-administration of multiple drugs can lead to alteration in the exposure of the victim drug, raising safety or effectiveness concerns. Assessment of the DDI potential starts with in vitro experiments to determine kinetic parameters and identify risks associated with the use of comedication that can inform future clinical studies. The diverse range of experimental models and techniques has significantly contributed to the examination of potential DDIs. Cytochrome P450 (CYP) enzymes are responsible for the biotransformation of many drugs on the market, making them frequently implicated in drug metabolism and DDIs. Consequently, there has been a growing focus on the assessment of DDI risk for CYPs. This review article provides mechanistic insights underlying CYP inhibition/induction and an overview of the in vitro assessment of CYP-mediated DDIs.
Collapse
Affiliation(s)
- Jonghwa Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.L.B.); (R.M.G.)
| | | | | | - Klarissa D. Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.L.B.); (R.M.G.)
| |
Collapse
|
10
|
Zhou Y, Dong H, Fan J, Zhu M, Liu L, Wang Y, Tang P, Chen X. Cytochrome P450 2B6 and UDP-Glucuronosyltransferase Enzyme-Mediated Clearance of Ciprofol (HSK3486) in Humans: The Role of Hepatic and Extrahepatic Metabolism. Drug Metab Dispos 2024; 52:106-117. [PMID: 38071562 DOI: 10.1124/dmd.123.001484] [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: 08/02/2023] [Revised: 11/01/2023] [Accepted: 12/06/2023] [Indexed: 12/22/2023] Open
Abstract
Ciprofol (HSK3486) is a novel intravenous agent for general anesthesia. In humans, HSK3486 mainly undergoes glucuronidation to form M4 [fraction of clearance (fCL): 62.6%], followed by the formation of monohydroxylated metabolites that further undergo glucuronidation and sulfation to produce M5-1, M5-2, M5-3, and M3 (summed fCL: 35.2%). However, the complete metabolic pathways of HSK3486 in humans remain unclear. In this study, by comparison with chemically synthesized reference standards, three monohydroxylated metabolites [M7-1, 4-hydroxylation with an unbound intrinsic clearance (CLint,u) of 2211 μl/min/mg; M7-2, ω-hydroxylation with a CLint,u of 600 μl/min/mg; and M7-3, (ω-1)-hydroxylation with a CLint,u of 78.4 μl/min/mg] were identified in human liver microsomes, and CYP2B6 primarily catalyzed their formation. In humans, M7-1 was shown to undergo glucuronidation at the 4-position and 1-position by multiple UDP-glucuronosyltransferases (UGTs) to produce M5-1 and M5-3, respectively, or was metabolized to M3 by cytosolic sulfotransferases. M7-2 was glucuronidated at the ω position by UGT1A9, 2B4, and 2B7 to form M5-2. UGT1A9 predominantly catalyzed the glucuronidation of HSK3486 (M4). The CLint,u values for M4 formation in human liver and kidney microsomes were 1028 and 3407 μl/min/mg, respectively. In vitro to in vivo extrapolation analysis suggested that renal glucuronidation contributed approximately 31.4% of the combined clearance. In addition to HSK3486 glucuronidation (M4), 4-hydroxylation (M7-1) was identified as another crucial oxidative metabolic pathway (fCL: 34.5%). Further attention should be paid to the impact of CYP2B6- and UGT1A9-mediated drug interactions and gene polymorphisms on the exposure and efficacy of HSK3486. SIGNIFICANCE STATEMENT: This research elucidates the major oxidative metabolic pathways of HSK3486 (the formation of three monohydroxylated metabolites: M7-1, M7-2, M7-3) as well as definitive structures and formation pathways of these monohydroxylated metabolites and their glucuronides or sulfate in humans. This research also identifies major metabolizing enzymes responsible for the glucuronidation (UGT1A9) and oxidation (CYP2B6) of HSK3486 and characterizes the mechanism of extrahepatic metabolism. The above information is helpful in guiding the safe use of HSK3486 in the clinic.
Collapse
Affiliation(s)
- Yufan Zhou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Hongjiao Dong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Jiang Fan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Mingshe Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Lu Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Yongbin Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Pingming Tang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Y.Z., L.L., Y.W., X.C.); University of Chinese Academy of Sciences, Beijing, China (Y.Z., X.C.); Haisco Pharmaceutical Group Co., Ltd., Chengdu, Sichuan Province, China (H.D., J.F., M.Z., P.T.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| |
Collapse
|
11
|
Furlani IL, Oliveira RV, Cass QB. Immobilization of cytochrome P450 enzymes onto magnetic beads: an approach to drug metabolism and biocatalysis. TALANTA OPEN 2023. [DOI: 10.1016/j.talo.2023.100181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|