1
|
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: 0] [Impact Index Per Article: 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
|
2
|
Podoll T, Pearson PG, Kaptein A, Evarts J, de Bruin G, Emmelot-van Hoek M, de Jong A, van Lith B, Sun H, Byard S, Fretland A, Hoogenboom N, Barf T, Slatter JG. Identification and Characterization of ACP-5862, the Major Circulating Active Metabolite of Acalabrutinib: Both Are Potent and Selective Covalent Bruton Tyrosine Kinase Inhibitors . J Pharmacol Exp Ther 2023; 384:173-186. [PMID: 36310034 DOI: 10.1124/jpet.122.001116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 12/27/2022] Open
Abstract
Acalabrutinib is a covalent Bruton tyrosine kinase (BTK) inhibitor approved for relapsed/refractory mantle cell lymphoma and chronic lymphocytic leukemia/small lymphocytic lymphoma. A major metabolite of acalabrutinib (M27, ACP-5862) was observed in human plasma circulation. Subsequently, the metabolite was purified from an in vitro biosynthetic reaction and shown by nuclear magnetic resonance spectroscopy to be a pyrrolidine ring-opened ketone/amide. Synthesis confirmed its structure, and covalent inhibition of wild-type BTK was observed in a biochemical kinase assay. A twofold lower potency than acalabrutinib was observed but with similar high kinase selectivity. Like acalabrutinib, ACP-5862 was the most selective toward BTK relative to ibrutinib and zanubrutinib. Because of the potency, ACP-5862 covalent binding properties, and potential contribution to clinical efficacy of acalabrutinib, factors influencing acalabrutinib clearance and ACP-5862 formation and clearance were assessed. rCYP (recombinant cytochrome P450) reaction phenotyping indicated that CYP3A4 was responsible for ACP-5862 formation and metabolism. ACP-5862 formation Km (Michaelis constant) and Vmax were 2.78 μM and 4.13 pmol/pmol CYP3A/min, respectively. ACP-5862 intrinsic clearance was 23.6 μL/min per mg. Acalabrutinib weakly inhibited CYP2C8, CYP2C9, and CYP3A4, and ACP-5862 weakly inhibited CYP2C9 and CYP2C19; other cytochrome P450s, UGTs (uridine 5'-diphospho-glucuronosyltransferases), and aldehyde oxidase were not inhibited. Neither parent nor ACP-5862 strongly induced CYP1A2, CYP2B6, or CYP3A4 mRNA. Acalabrutinib and ACP-5862 were substrates of multidrug resistance protein 1 and breast cancer resistance protein but not OATP1B1 or OATP1B3. Our work indicates that ACP-5862 may contribute to clinical efficacy in acalabrutinib-treated patients and illustrates how proactive metabolite characterization allows timely assessment of drug-drug interactions and potential contributions of metabolites to pharmacological activity. SIGNIFICANCE STATEMENT: This work characterized the major metabolite of acalabrutinib, ACP-5862. Its contribution to the pharmacological activity of acalabrutinib was assessed based on covalent Bruton tyrosine kinase binding kinetics, kinase selectivity, and potency in cellular assays. The metabolic clearance and in vitro drug-drug interaction potential were also evaluated for both acalabrutinib and ACP-5862. The current data suggest that ACP-5862 may contribute to the clinical efficacy observed in acalabrutinib-treated patients and demonstrates the value of proactive metabolite identification and pharmacological characterization.
Collapse
Affiliation(s)
- Terry Podoll
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Paul G Pearson
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Allard Kaptein
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Jerry Evarts
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Gerjan de Bruin
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Maaike Emmelot-van Hoek
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Anouk de Jong
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Bart van Lith
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Hao Sun
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Stephen Byard
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Adrian Fretland
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Niels Hoogenboom
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - Tjeerd Barf
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| | - J Greg Slatter
- Acerta Pharma (a member of the AstraZeneca group), South San Francisco, California (T.P., J.E., A.F., J.G.S.); Acerta Pharma (a member of the AstraZeneca group) Oss, The Netherlands (G.d.B., M.E.-v.H., A.d.J., B.v.L., N.H.); Pearson Pharma Partners, Westlake Village, California (P.G.P.); Covance Laboratories, Madison, Wisconsin (H.S.); Arcinova, Alnwick, United Kingdom (S.B.); and Covalution Holding B.V., Ravenstein, The Netherlands (A.K., T.B.)
| |
Collapse
|
3
|
Lin F, Lin X, Wang X, Mei G, Chen B, Yao H, Huang L. Inhibitory effect of Selaginella doederleinii hieron on human cytochrome P450. Front Pharmacol 2023; 14:1108867. [PMID: 36874034 PMCID: PMC9975586 DOI: 10.3389/fphar.2023.1108867] [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: 11/26/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Introduction: Selaginella doederleinii Hieron is a traditional Chinese herbal medicine, the ethyl acetate extract from Selaginella doederleinii (SDEA) showed favorable anticancer potentials. However, the effect of SDEA on human cytochrome P450 enzymes (CYP450) remains unclear. To predict the herb-drug interaction (HDI) and lay the groundwork for further clinical trials, the inhibitory effect of SDEA and its four constituents (Amentoflavone, Palmatine, Apigenin, Delicaflavone) on seven CYP450 isoforms were investigated by using the established CYP450 cocktail assay based on LC-MS/MS. Methods: Appropriate substrates for seven tested CYP450 isoforms were selected to establish a reliable cocktail CYP450 assay based on LC-MS/MS. The contents of four constituents (Amentoflavone, Palmatine, Apigenin, Delicaflavone) in SDEA were determined as well. Then, the validated CYP450 cocktail assay was applied to test the inhibitory potential of SDEA and four constituents on CYP450 isoforms. Results: SDEA showed strong inhibitory effect on CYP2C9 and CYP2C8 (IC50 ≈ 1 μg/ml), moderate inhibitory effect against CYP2C19, CYP2E1 and CYP3A (IC50 < 10 μg/ml). Among the four constituents, Amentoflavone had the highest content in the extract (13.65%) and strongest inhibitory effect (IC50 < 5 μM), especially for CYP2C9, CYP2C8 and CYP3A. Amentoflavone also showed time-dependent inhibition on CYP2C19 and CYP2D6. Apigenin and Palmatine both showed concentration-dependent inhibition. Apigenin inhibited CYP1A2, CYP2C8, CYP2C9, CYP2E1 and CYP3A. Palmatine inhibited CYP3A and had a weak inhibitory effect on CYP2E1. As for Delicaflavone, which has the potential to develop as an anti-cancer agent, showed no obvious inhibitory effect on CYP450 enzymes. Conclusion: Amentoflavone may be one of the main reasons for the inhibition of SDEA on CYP450 enzymes, the potential HDI should be considered when SDEA or Amentoflavone were used with other clinical drugs. On the contrast, Delicaflavone is more suitable to develop as a drug for clinical use, considering the low level of CYP450 metabolic inhibition.
Collapse
Affiliation(s)
- Fei Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xinhua Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xuewen Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Guanghui Mei
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bing Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Hong Yao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Lingyi Huang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
4
|
Tang LWT, Fu J, Koh SK, Wu G, Zhou L, Chan ECY. Metabolic Activation of the Acrylamide Michael Acceptor Warhead in Futibatinib to an Epoxide Intermediate Engenders Covalent Inactivation of Cytochrome P450 3A. Drug Metab Dispos 2022; 50:931-941. [PMID: 35512804 DOI: 10.1124/dmd.122.000895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
Futibatinib (FUT) is a potent inhibitor of fibroblast growth factor receptor (FGFR) 1-4 that is currently under clinical investigation for intrahepatic cholangiocarcinoma. Unlike its predecessors, FUT possesses an acrylamide warhead which enables it to bind covalently to a free cysteine residue in the FGFR kinase domain. However, it remains uninterrogated if this electrophilic α,β-unsaturated carbonyl scaffold could also directly or indirectly engender off-target covalent binding to nucleophilic centres on other cellular proteins. Here, we discovered that FUT inactivated both cytochrome P450 3A (CYP3A) isoforms with K I, k inact, and partition ratio of 12.5 and 51.4 µM, 0.25 and 0.06 min-1 and ~52 and ~58 for CYP3A4 and CYP3A5, respectively. Along with its time-, concentration- and cofactor-dependent inhibitory profile, FUT also exhibited several cardinal features that were consistent with mechanism-based inactivation. Moreover, the nature of inactivation was unlikely to be pseudo-irreversible and instead arose from the covalent modification of the P450 apoprotein and/or its heme moiety due to the lack of substantial enzyme activity recovery following dialysis and chemical oxidation as well as the absence of the diagnostic Soret peak in spectral analyses. Finally, utilizing GSH trapping and high-resolution mass spectrometry, we illuminated that while the acrylamide moiety in FUT could nonenzymatically conjugate to GSH via Michael addition, it was not implicated in the covalent inactivation of CYP3A. Rather, we surmised that it likely stemmed from the metabolic activation of its acrylamide covalent warhead to a highly electrophilic epoxide intermediate that could covalently modify CYP3A and culminate in its catalytic inactivation. Significance Statement In this study, we reported for the first time the inactivation of CYP3A by FUT. Furthermore, using FUT as an exemplary targeted covalent inhibitor, our study revealed the propensity for its acrylamide Michael acceptor moiety to be metabolically activated to a highly electrophilic epoxide. Due to the growing resurgence of covalent inhibitors and the well-established toxicological ramifications associated with epoxides, we advocate that closer scrutiny be adopted when profiling the reactive metabolites of compounds possessing an α,β-unsaturated carbonyl scaffold.
Collapse
Affiliation(s)
| | - Jiaxin Fu
- National University of Singapore, Singapore
| | | | - Guoyi Wu
- National University of Singapore, Singapore
| | - Lei Zhou
- Singapore Eye Research Institute, Singapore
| | | |
Collapse
|
5
|
Tang LWT, Wei W, Verma RK, Koh SK, Zhou L, Fan H, Chan ECY. Direct and Sequential Bioactivation of Pemigatinib to Reactive Iminium Ion Intermediates Culminate in Mechanism-Based Inactivation of Cytochrome P450 3A. Drug Metab Dispos 2022; 50:529-540. [DOI: 10.1124/dmd.121.000804] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/01/2022] [Indexed: 11/22/2022] Open
|
6
|
Tu D, Ning J, Zou L, Wang P, Zhang Y, Tian X, Zhang F, Zheng J, Ge G. Unique Oxidative Metabolism of Bufalin Generates Two Reactive Metabolites That Strongly Inactivate Human Cytochrome P450 3A. J Med Chem 2022; 65:4018-4029. [DOI: 10.1021/acs.jmedchem.1c01875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Dongzhu Tu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Ning
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Liwei Zou
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yani Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangge Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550025, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
7
|
Ning J, Tian Z, Wang J, Wang B, Tian X, Yu Z, Huo X, Feng L, Cui J, James TD, Ma X. Rational Design of a Two‐Photon Fluorescent Probe for Human Cytochrome P450 3A and the Visualization of Mechanism‐Based Inactivation. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202113191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jing Ning
- Second Affiliated Hospital of Dalian Medical University Dalian 116023 China
- College of Integrative Medicine College of Pharmacy Dalian Medical University Dalian 116044 China
| | - Zhenhao Tian
- School of Life Sciences Northwestern Polytechnical University Xi'an 710072 China
| | - Jiayue Wang
- College of Integrative Medicine College of Pharmacy Dalian Medical University Dalian 116044 China
- Department of Pharmacy Peking University Shenzhen Hospital Shenzhen 518036 China
| | - Bo Wang
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing China
| | - Xiangge Tian
- College of Integrative Medicine College of Pharmacy Dalian Medical University Dalian 116044 China
| | - Zhenlong Yu
- College of Integrative Medicine College of Pharmacy Dalian Medical University Dalian 116044 China
| | - Xiaokui Huo
- College of Integrative Medicine College of Pharmacy Dalian Medical University Dalian 116044 China
| | - Lei Feng
- Second Affiliated Hospital of Dalian Medical University Dalian 116023 China
- School of Chemistry and Chemical Engineering Henan Normal University Xinxiang 453007 China
| | - Jingnan Cui
- State Key Laboratory of Fine Chemicals Dalian University of Technology Dalian 116024 China
| | - Tony D. James
- School of Chemistry and Chemical Engineering Henan Normal University Xinxiang 453007 China
- Department of Chemistry University of Bath Bath BA2 7AY United Kingdom
| | - Xiaochi Ma
- Second Affiliated Hospital of Dalian Medical University Dalian 116023 China
- College of Integrative Medicine College of Pharmacy Dalian Medical University Dalian 116044 China
| |
Collapse
|
8
|
Ning J, Tian Z, Wang J, Wang B, Tian X, Yu Z, Huo X, Feng L, Cui J, James TD, Ma X. Rational Design of a Two-Photon Fluorescent Probe for Human Cytochrome P450 3A and the Visualization of Mechanism-Based Inactivation. Angew Chem Int Ed Engl 2022; 61:e202113191. [PMID: 34851011 DOI: 10.1002/anie.202113191] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Indexed: 12/18/2022]
Abstract
Mechanism-based inactivation (MBI) can mediate adverse reactions and hepatotoxicity from drugs, which is a result of their conversion into highly reactive metabolites catalyzed by enzymes such as cytochrome P450 3A (CYP3A). In the present research, we optimized the key interaction domain of the fluorophore with the target protein to develop a two-photon fluorescent probe for CYP3A that is involved in the metabolism of more than half of all clinical drugs. The developed BN-1 probe exhibited appropriate selectivity and sensitivity for the semi-quantitative detection and imaging of endogenous CYP3A activity in various living systems, thereby providing a high-throughput screening system enabling evaluation of MBI-associated hepatotoxicity by CYP3A. Using BN-1 as a fluorescent molecular tool facilitates the efficient discovery and characterization of CYP3A-induced MBI in natural systems.
Collapse
Affiliation(s)
- Jing Ning
- Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zhenhao Tian
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jiayue Wang
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Bo Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiangge Tian
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zhenlong Yu
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xiaokui Huo
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Lei Feng
- Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China
| | - Jingnan Cui
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, 116024, China
| | - Tony D James
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China
- Department of Chemistry, University of Bath, Bath, BA2 7AY, United Kingdom
| | - Xiaochi Ma
- Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|
9
|
Toda N, Shida T, Takano R, Katagiri T, Hirouchi M, Abe M, Soma K, Nakagami Y, Yamazaki M. Discovery of DS-3801b, a non-macrolide GPR38 agonist with N-methylanilide structure. Bioorg Med Chem Lett 2022; 59:128554. [DOI: 10.1016/j.bmcl.2022.128554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 11/17/2022]
|
10
|
Tang LWT, Teng JW, Verma RK, Koh SK, Zhou L, Go ML, Fan H, Chan ECY. Infigratinib is a Reversible Inhibitor and Mechanism-based Inactivator of Cytochrome P450 3A4. Drug Metab Dispos 2021; 49:856-868. [PMID: 34326139 DOI: 10.1124/dmd.121.000508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/08/2021] [Indexed: 11/22/2022] Open
Abstract
Infigratinib (INF) is a promising selective inhibitor of fibroblast growth factor receptors 1-3 that has recently been accorded both orphan drug designation and priority review status by the U.S Food and Drug Administration for the treatment of advanced cholangiocarcinoma. Its propensity to undergo bioactivation to electrophilic species was recently expounded upon. However, other than causing aberrant idiosyncratic toxicities, these reactive intermediates may elicit mechanism-based inactivation (MBI) of cytochrome P450 enzymes (CYP450). In this study, we investigated the interactions between INF and the most abundant hepatic cytochrome P450 3A4 (CYP3A4). Our findings revealed that apart from being a potent noncompetitive reversible inhibitor of CYP3A4, INF inactivated CYP3A4 in a time-, concentration- and NADPH-dependent manner with K I, k inact and partition ratio of 2.45 µM, 0.053 min-1 and 41 respectively when rivaroxaban was employed as the probe substrate. Co-incubation with testosterone (alternative CYP3A substrate) or ketoconazole (direct CYP3A inhibitor) attenuated the rate of inactivation whereas the inclusion of glutathione and catalase did not confer such protection. The lack of enzyme activity recovery following dialysis for 4 hours and oxidation with potassium ferricyanide, coupled with the absence of the characteristic Soret peak signature collectively substantiated that inactivation of CYP3A4 by INF was not mediated by the formation of quasi-irreversible metabolite-intermediate complexes but rather through irreversible covalent adduction to the prosthetic heme and/or apoprotein. Finally, glutathione trapping and high-resolution mass spectrometry experimental results unravelled two plausible bioactivation mechanisms of INF arising from the generation of a p-benzoquinone diimine and epoxide reactive intermediate. Significance Statement The potential of infigratinib (INF) to cause mechanism-based inactivation (MBI) of CYP3A4 was unknown. We report the reversible noncompetitive inhibition and irreversible covalent MBI of CYP3A4 by INF and proposed two potential bioactivation pathways implicating p-benzoquinone diimine and epoxide reactive intermediates. Findings from this study lay the groundwork for future investigation of clinically-relevant drug-drug interactions between INF and concomitant substrates of CYP3A4.
Collapse
Affiliation(s)
| | | | - Ravi Kumar Verma
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
| | | | - Lei Zhou
- Singapore Eye Research Institute, Singapore
| | - Mei Lin Go
- National University of Singapore, Singapore
| | - Hao Fan
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
11
|
Tang LWT, Teng JW, Koh SK, Zhou L, Go ML, Chan ECY. Mechanism-Based Inactivation of Cytochrome P450 3A4 and 3A5 by the Fibroblast Growth Factor Receptor Inhibitor Erdafitinib. Chem Res Toxicol 2021; 34:1800-1813. [PMID: 34189909 DOI: 10.1021/acs.chemrestox.1c00178] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Erdafitinib (ERD) is a first-in-class pan inhibitor of fibroblast growth factor receptor 1-4 that has garnered global regulatory approval for the treatment of advanced or metastatic urothelial carcinoma. Although it has been previously reported that ERD elicits time-dependent inhibition (TDI) of cytochrome P450 (P450) 3A4 (CYP3A4), the exact biochemical nature underpinning this observation remains obfuscated. Moreover, it is also uninterrogated if CYP3A5-its highly homologous counterpart-could be susceptible to such interactions. Mechanism-based inactivation (MBI) of P450 is a unique subset of TDI that hinges on prior bioactivation of the drug to a reactive intermediate and possesses profound clinical and toxicological implications due to its irreversible nature. Here, we investigated and confirmed that ERD inactivated both CYP3A isoforms in a time-, concentration-, and NADPH-dependent manner with KI, kinact, and partition ratio of 4.01 and 10.04 μM, 0.120 and 0.045 min-1, and 32 and 55 for both CYP3A4 and CYP3A5, respectively, when rivaroxaban was employed as the probe substrate. Co-incubation with an alternative substrate or direct inhibitor of CYP3A attenuated the rate of inactivation, whereas the addition of glutathione or catalase did not induce such protection. The lack of enzyme activity recovery following dialysis for 4 h and oxidation with potassium ferricyanide combined with the lack of a Soret peak in spectral scans collectively substantiated that ERD is an irreversible covalent MBI of CYP3A. Finally, glutathione trapping and high-resolution mass spectrometry experiments illuminated a plausible bioactivation mechanism of ERD by CYP3A arising from metabolic epoxidation of its quinoxaline ring.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Department of Pharmacy, Faculty of Science, National University of Singapore, 169856 Singapore
| | - Jian Wei Teng
- Department of Pharmacy, Faculty of Science, National University of Singapore, 169856 Singapore
| | | | - Lei Zhou
- Singapore Eye Research Institute (SERI), Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore.,Ophthalmology and Visual Sciences Academia Clinical Program, Duke-National University of Singapore Medical School, 169857 Singapore
| | - Mei Lin Go
- Department of Pharmacy, Faculty of Science, National University of Singapore, 169856 Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 169856 Singapore
| |
Collapse
|
12
|
Tu DZ, Mao X, Zhang F, He RJ, Wu JJ, Wu Y, Zhao XH, Zheng J, Ge GB. Reversible and Irreversible Inhibition of Cytochrome P450 Enzymes by Methylophiopogonanone A. Drug Metab Dispos 2021; 49:459-469. [PMID: 33811108 DOI: 10.1124/dmd.120.000325] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/09/2021] [Indexed: 01/07/2023] Open
Abstract
Methylophiopogonanone A (MOA), an abundant homoisoflavonoid bearing a methylenedioxyphenyl moiety, is one of the major constituents in the Chinese herb Ophiopogon japonicas This work aims to assess the inhibitory potentials of MOA against cytochrome P450 enzymes and to decipher the molecular mechanisms for P450 inhibition by MOA. The results showed that MOA concentration-dependently inhibited CYP1A, 2C8, 2C9, 2C19, and 3A in human liver microsomes (HLMs) in a reversible way, with IC50 values varying from 1.06 to 3.43 μM. By contrast, MOA time-, concentration-, and NADPH-dependently inhibited CYP2D6 and CYP2E1, along with KI and kinact values of 207 µM and 0.07 minute-1 for CYP2D6, as well as 20.9 µM and 0.03 minutes-1 for CYP2E1. Further investigations demonstrated that a quinone metabolite of MOA could be trapped by glutathione in an HLM incubation system, and CYP2D6, 1A2, and 2E1 were the major contributors to catalyze the metabolic activation of MOA to the corresponding O-quinone intermediate. Additionally, the potential risks of herb-drug interactions triggered by MOA or MOA-related products were also predicted. Collectively, our findings verify that MOA is a reversible inhibitor of CYP1A, 2C8, 2C9, 2C19, and 3A but acts as an inactivator of CYP2D6 and CYP2E1. SIGNIFICANCE STATEMENT: Methylophiopogonanone A (MOA), an abundant homoisoflavonoid isolated from the Chinese herb Ophiopogon japonicas, is a reversible inhibitor of CYP1A, 2C8, 2C9, 2C19, and 3A but acts as an inactivator of CYP2D6 and CYP2E1. Further investigations demonstrated that a quinone metabolite of MOA could be trapped by glutathione in a human liver microsome incubation system, and CYP2D6, 1A2, and 2E1 were the major contributors to catalyze the metabolic activation of MOA to the corresponding O-quinone intermediate.
Collapse
Affiliation(s)
- Dong-Zhu Tu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Xu Mao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Feng Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Rong-Jing He
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Jing-Jing Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Yue Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Xiao-Hua Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Jiang Zheng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| |
Collapse
|
13
|
Tang LWT, Verma RK, Fan H, Chan ECY. Mechanism-Based Inactivation of Cytochrome P450 3A4 by Benzbromarone. Mol Pharmacol 2021; 99:266-276. [PMID: 33436520 DOI: 10.1124/molpharm.120.000086] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
Benzbromarone (BBR), a potent uricosuric agent for the management of gout, is known to cause fatal fulminant hepatitis. Although the mechanism of BBR-induced idiosyncratic hepatotoxicity remains unelucidated, cytochrome P450 enzyme-mediated bioactivation of BBR to electrophilic reactive metabolites is commonly regarded as a key molecular initiating event. However, apart from causing aberrant toxicities, reactive metabolites may result in mechanism-based inactivation (MBI) of cytochrome P450. Here, we investigated and confirmed that BBR inactivated CYP3A4 in a time-, concentration-, and NADPH-dependent manner with K I, k inact, and partition ratio of 11.61 µM, 0.10 minutes-1, and 110, respectively. Coincubation with ketoconazole, a competitive inhibitor of CYP3A4, attenuated the MBI of CYP3A4 by BBR, whereas the presence of glutathione and catalase did not confer such protection. The lack of substantial recovery of enzyme activity postdialysis and after oxidation with potassium ferricyanide, combined with the absence of a Soret peak in spectral difference scans, implied that MBI of CYP3A4 by BBR did not occur through the formation of quasi-irreversible metabolite-intermediate complexes. Analysis of the reduced CO-difference spectrum revealed an ∼44% reduction in ferrous-CO binding and hinted that inactivation is mediated via irreversible covalent adduction to both the prosthetic heme moiety and the apoprotein. Finally, our in silico covalent docking analysis further suggested the modulation of substrate binding to CYP3A4 via the covalent adduction of epoxide-derived reactive intermediates of BBR to two key cysteine residues (Cys239 and Cys58) vicinal to the entrance of the orthosteric binding site. SIGNIFICANCE STATEMENT: Although the bioactivation of benzbromarone (BBR) to reactive metabolites has been well characterized, its potential to cause mechanism-based inactivation (MBI) of cytochrome P450 has not been fully investigated. This study reports the MBI of CYP3A4 by BBR via irreversible covalent adduction and develops a unique covalent docking methodology to predict the structural molecular determinants underpinning the inactivation for the first time. These findings lay the groundwork for future investigation of clinically relevant drug-drug interactions implicating BBR and mechanisms of BBR-induced idiosyncratic hepatotoxicity.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore (L.W.T.T., E.C.Y.C.) and Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore (R.K.V., H.F.)
| | - Ravi Kumar Verma
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore (L.W.T.T., E.C.Y.C.) and Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore (R.K.V., H.F.)
| | - Hao Fan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore (L.W.T.T., E.C.Y.C.) and Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore (R.K.V., H.F.)
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore (L.W.T.T., E.C.Y.C.) and Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore (R.K.V., H.F.)
| |
Collapse
|
14
|
Li H, Tang Y, Wei W, Yin C, Tang F. Effects of saikosaponin-d on CYP3A4 in HepaRG cell and protein-ligand docking study. Basic Clin Pharmacol Toxicol 2020; 128:661-668. [PMID: 33369126 DOI: 10.1111/bcpt.13552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/22/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023]
Abstract
Saikosaponin-d (SSd) is a major bioactive triterpenoid saponin extracted from Bupleurum, which has anti-inflammatory, anticancer, antioxidative and anti-hepatic fibrosis effects. Due to the effects of Bupleurum-related formulations on cytochrome P450 (CYPs) expression still remain unclear, the combination therapies involved formulations containing Bupleurum may sometimes lead to unexpected drug-drug interactions in clinical practice. These interactions can limit the clinical applications of related formulations. In this study, we tried to explore the effects of SSd on CYP3A4 mRNA, protein expression and the enzyme activity in HepaRG cells by real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR), Western blot (WB) and HPLC method, respectively. The interaction between SSd and CYP3A4 was analysed by molecular docking. HepaRG cells were cultured with different concentrations of SSd (0.5, 1, 5 and 10 μmol/L) for 72 hours. It is revealed that SSd can inhibit CYP3A4 mRNA and its protein expression, and also the enzyme activity. Molecular docking study demonstrated that SSd can bind to several key active sites of amino acid residues of CYP3A4 protein with hydrogen bonds and hydrophobic interactions. Thus, drug-drug interactions resulted by SSd inhibiting CYP3A4 need attention when formulations containing SSd or Bupleurum are co-administrated with drugs metabolized by CYP3A4.
Collapse
Affiliation(s)
- Hongfang Li
- Department of Clinical Pharamcy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| | - Yunyan Tang
- Department of Clinical Pharamcy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China.,Department of Pharmacy, Meitan People's Hospital, Zunyi, China
| | - Weipeng Wei
- Department of Clinical Pharamcy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| | - Chengchen Yin
- Department of Clinical Pharamcy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| | - Fushan Tang
- Department of Clinical Pharamcy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Clinical Pharmacy of Zunyi City, Zunyi Medical University, Zunyi, China
| |
Collapse
|
15
|
Shinozuka T, Kobayashi H, Suzuki S, Tanaka K, Karanjule N, Hayashi N, Tsuda T, Tokumaru E, Inoue M, Ueda K, Kimoto H, Domon Y, Takahashi S, Kubota K, Yokoyama T, Shimizugawa A, Koishi R, Fujiwara C, Asano D, Sakakura T, Takasuna K, Abe Y, Watanabe T, Kitano Y. Discovery of DS-1971a, a Potent, Selective NaV1.7 Inhibitor. J Med Chem 2020; 63:10204-10220. [DOI: 10.1021/acs.jmedchem.0c00259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hiroyuki Kobayashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Sayaka Suzuki
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kyosuke Tanaka
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Narayan Karanjule
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Noriyuki Hayashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Toshifumi Tsuda
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Eri Tokumaru
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masahiro Inoue
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kiyono Ueda
- R&D Division, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Hiroko Kimoto
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yuki Domon
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Sakiko Takahashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kazufumi Kubota
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Tomihisa Yokoyama
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Akiko Shimizugawa
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Ryuta Koishi
- R&D Division, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Chie Fujiwara
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Daigo Asano
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Tomoko Sakakura
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kiyoshi Takasuna
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yasuyuki Abe
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Toshiyuki Watanabe
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
16
|
Yadav J, Paragas E, Korzekwa K, Nagar S. Time-dependent enzyme inactivation: Numerical analyses of in vitro data and prediction of drug-drug interactions. Pharmacol Ther 2020; 206:107449. [PMID: 31836452 PMCID: PMC6995442 DOI: 10.1016/j.pharmthera.2019.107449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cytochrome P450 (CYP) enzyme kinetics often do not conform to Michaelis-Menten assumptions, and time-dependent inactivation (TDI) of CYPs displays complexities such as multiple substrate binding, partial inactivation, quasi-irreversible inactivation, and sequential metabolism. Additionally, in vitro experimental issues such as lipid partitioning, enzyme concentrations, and inactivator depletion can further complicate the parameterization of in vitro TDI. The traditional replot method used to analyze in vitro TDI datasets is unable to handle complexities in CYP kinetics, and numerical approaches using ordinary differential equations of the kinetic schemes offer several advantages. Improvement in the parameterization of CYP in vitro kinetics has the potential to improve prediction of clinical drug-drug interactions (DDIs). This manuscript discusses various complexities in TDI kinetics of CYPs, and numerical approaches to model these complexities. The extrapolation of CYP in vitro TDI parameters to predict in vivo DDIs with static and dynamic modeling is discussed, along with a discussion on current gaps in knowledge and future directions to improve the prediction of DDI with in vitro data for CYP catalyzed drug metabolism.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Amgen Inc., 360 Binney Street, Cambridge, MA 02142, United States; Department of Pharmaceutical Sciences, Temple University, Philadelphia, PA 19140, United States
| | - Erickson Paragas
- Department of Pharmaceutical Sciences, Temple University, Philadelphia, PA 19140, United States
| | - Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University, Philadelphia, PA 19140, United States
| | - Swati Nagar
- Department of Pharmaceutical Sciences, Temple University, Philadelphia, PA 19140, United States.
| |
Collapse
|
17
|
Lu C, Di L. In vitro
and
in vivo
methods to assess pharmacokinetic drug– drug interactions in drug discovery and development. Biopharm Drug Dispos 2020; 41:3-31. [DOI: 10.1002/bdd.2212] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/27/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Chuang Lu
- Department of DMPKSanofi Company Waltham MA 02451
| | - Li Di
- Pharmacokinetics, Dynamics and MetabolismPfizer Worldwide Research & Development Groton CT 06340
| |
Collapse
|
18
|
Cui T, Wang Q, Tian X, Zhang K, Peng Y, Zheng J. Piperine Is a Mechanism-Based Inactivator of CYP3A. Drug Metab Dispos 2019; 48:123-134. [PMID: 31748224 DOI: 10.1124/dmd.119.088955] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/05/2019] [Indexed: 11/22/2022] Open
Abstract
Piperine (PPR) is the representative alkaloid component of the piper species (family: Piperaceae). Our rapid screening study found PPR caused time-dependent inhibition of cytochrome P450s (CYP) 3A and 2D6, and CYP3A was inactivated the most. Further study demonstrated that PPR is a time-, concentration-, and NADPH-dependent inhibitor of CYP3A, and significant loss (49.5% ± 3.9%) of CYP3A activity was observed after 20minute incubations with 80 μM PPR at 37°C. The values of K I and k inact were 30.7 μM and 0.041 minutes-1, respectively. CYP3A competitive inhibitor ketoconazole showed protective effect against the enzyme inactivation. Superoxide dismutase/catalase and GSH displayed minor protection against the PPR-caused enzyme inactivation. Ferricyanide partially reduced the enzyme inhibition by PPR. Additionally, NADPH-dependent formation of reactive metabolites from PPR were found in human liver microsomal incubation mixtures. An ortho-quinone intermediate was trapped by NAC in microsomal incubations with PPR. DM-PPR, demethylene metabolite of PPR, showed weak enzyme inactivation relative to that caused by PPR. It appears that both carbene and ortho-quinone intermediates were involved in the inactivation of CYP3A caused by PPR. SIGNIFICANCE STATEMENT: CYP3A subfamily members (mainly CYP3A4 and CYP3A5) play a critical role in drug metabolism. Piperine (PPR), a methylenedioxyphenyl derivative combined with an unsaturated ketone, is the major active ingredient of pepper. PPR revealed time-, concentration-, and NADPH-dependent inhibitory effect on CYP3A. Carbene and quinone metabolites were both involved in the observed CYP3A inactivation by PPR. Apparently, the unsaturated ketone moiety did not participate in the enzyme inactivation. The present study sounds an alert of potential risk for food-drug interactions.
Collapse
Affiliation(s)
- Tiantian Cui
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, and Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, P. R. China(J.Z.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China(T.C., Q.W., X.T., K.Z., Y.P., J.Z.)
| | - Qian Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, and Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, P. R. China(J.Z.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China(T.C., Q.W., X.T., K.Z., Y.P., J.Z.)
| | - Xiaoxiao Tian
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, and Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, P. R. China(J.Z.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China(T.C., Q.W., X.T., K.Z., Y.P., J.Z.)
| | - Kehan Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, and Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, P. R. China(J.Z.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China(T.C., Q.W., X.T., K.Z., Y.P., J.Z.)
| | - Ying Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, and Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, P. R. China(J.Z.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China(T.C., Q.W., X.T., K.Z., Y.P., J.Z.)
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, and Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, P. R. China(J.Z.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China(T.C., Q.W., X.T., K.Z., Y.P., J.Z.)
| |
Collapse
|
19
|
Yadav J, Korzekwa K, Nagar S. Impact of Lipid Partitioning on the Design, Analysis, and Interpretation of Microsomal Time-Dependent Inactivation. Drug Metab Dispos 2019; 47:732-742. [PMID: 31043439 PMCID: PMC6556519 DOI: 10.1124/dmd.118.085969] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/30/2019] [Indexed: 12/20/2022] Open
Abstract
Nonspecific drug partitioning into microsomal membranes must be considered for in vitro-in vivo correlations. This work evaluated the effect of including lipid partitioning in the analysis of complex TDI kinetics with numerical methods. The covariance between lipid partitioning and multiple inhibitor binding was evaluated. Simulations were performed to test the impact of lipid partitioning on the interpretation of TDI kinetics, and experimental TDI datasets for paroxetine (PAR) and itraconazole (ITZ) were modeled. For most kinetic schemes, modeling lipid partitioning results in statistically better fits. For MM-IL simulations (KI,u = 0.1 µM, kinact = 0.1 minute-1), concurrent modeling of lipid partitioning for an fumic range (0.01, 0.1, and 0.5) resulted in better fits compared with post hoc correction (AICc: -526 vs. -496, -579 vs. -499, and -636 vs. -579, respectively). Similar results were obtained with EII-IL. Lipid partitioning may be misinterpreted as double binding, leading to incorrect parameter estimates. For the MM-IL datasets, when fumic = 0.02, MM-IL, and EII model fits were indistinguishable (δAICc = 3). For less partitioned datasets (fumic = 0.1 or 0.5), the inclusion of partitioning resulted in better models. The inclusion of lipid partitioning can lead to markedly different estimates of KI,u and kinact A reasonable alternate experimental design is nondilution TDI assays, with post hoc fumic incorporation. The best fit models for PAR (MIC-M-IL) and ITZ (MIC-EII-M-IL and MIC-EII-M-Seq-IL) were consistent with their reported mechanism and kinetics. Overall, experimental fumic values should be concurrently incorporated into TDI models with complex kinetics, when dilution protocols are used.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| | - Swati Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania
| |
Collapse
|
20
|
Liu Y, Cui T, Peng Y, Ji M, Zheng J. Mechanism-based inactivation of cytochrome P450 2D6 by chelidonine. J Biochem Mol Toxicol 2018; 33:e22251. [PMID: 30368994 DOI: 10.1002/jbt.22251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 09/19/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022]
Abstract
Chelidonine (CHE) is a major bioactive constituent of greater celandine, a plant used in traditional herbal medicines. CHE has widely been used as an analgesic in clinical settings. We evaluated the inhibitory effects of CHE on human cytochrome P450 enzymes. CHE produced time-, concentration-, and NADPH-dependent inhibition of CYP2D6, with K I and k inact values of 20.49 μM and 11.05 min -1 , respectively. Approximately 76% of CYP2D6 activity was suppressed after 9 minute incubation with CHE (50 μM). The loss of enzyme activity was not restored following dialysis. The estimated partition ratio of the inactivation was about 156. Quinidine, a competitive inhibitor of CYP2D6, attenuated the CHE-mediated enzyme inactivation, while glutathione and catalase/superoxide dismutase did not markedly ameliorate the inhibitory effect. Upon oxidation using potassium ferricyanide, the 15.1% activity of CYP2D6 was restored. These findings indicate that CHE acted as a mechanism-based inactivator of CYP2D6 and the observed effects may induce potential drug-drug interactions.
Collapse
Affiliation(s)
- Yuyang Liu
- College of Plant Protection, Shenyang Agricultural University, Liaoning, Shenyang, China
| | - Tiantian Cui
- Wuya College of Innovation, Shenyang Pharmaceutical University, Liaoning, Shenyang, China
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Liaoning, Shenyang, China
| | - Mingshan Ji
- College of Plant Protection, Shenyang Agricultural University, Liaoning, Shenyang, China
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.,Wuya College of Innovation, Shenyang Pharmaceutical University, Liaoning, Shenyang, China
| |
Collapse
|
21
|
Langan LM, Owen SF, Trznadel M, Dodd NJF, Jackson SK, Purcell WM, Jha AN. Spheroid Size Does not Impact Metabolism of the β-blocker Propranolol in 3D Intestinal Fish Model. Front Pharmacol 2018; 9:947. [PMID: 30186177 PMCID: PMC6113889 DOI: 10.3389/fphar.2018.00947] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/02/2018] [Indexed: 12/12/2022] Open
Abstract
Compared to two-dimensional (2D) cell culture, cellular aggregates or spheroids (3D) offer a more appropriate alternative in vitro system where individual cell-cell communication and micro-environment more closely represent the in vivo organ; yet we understand little of the physiological conditions at this scale. The relationship between spheroid size and oxygen microenvironment, an important factor influencing the metabolic capacity of cells, was first established using the fish intestine derived RTgutGC cell line. Subsequently, pharmaceutical metabolism (Propranolol), as determined by high performance liquid chromatography, in this intestinal model was examined as a function of spheroid size. Co-efficient of variation between spheroid size was below 12% using the gyratory platform method, with the least variation observed in the highest cell seeding density. The viable, high oxygen micro-environment of the outer rim of the spheroid, as determined by electron paramagnetic resonance (EPR) oximetry, decreased over time, and the hypoxic zone increased as a function of spheroid size. Despite a trend of higher metabolism in smaller spheroids, the formation of micro-environments (quiescent, hypoxic or anoxic) did not significantly affect metabolism or function of an environmentally relevant pharmaceutical in this spheroid model.
Collapse
Affiliation(s)
- Laura M Langan
- School of Biological and Marine Sciences, University of Plymouth, Plymouth, United Kingdom
| | - Stewart F Owen
- Global Sustainability, AstraZeneca, Macclesfield, United Kingdom
| | - Maciej Trznadel
- College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Nicholas J F Dodd
- School of Biomedical and Healthcare Sciences, University of Plymouth, Plymouth, United Kingdom
| | - Simon K Jackson
- School of Biomedical and Healthcare Sciences, University of Plymouth, Plymouth, United Kingdom
| | - Wendy M Purcell
- Harvard T.H. Chan School of Public Health, Harvard University, Cambridge, MA, United States
| | - Awadhesh N Jha
- School of Biological and Marine Sciences, University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
22
|
Odagiri T, Inagaki H, Nagamochi M, Kitamura T, Komoriya S, Takahashi H. Design, Synthesis, and Biological Evaluation of Novel 7-[(3aS,7aS)-3a-Aminohexahydropyrano[3,4-c]pyrrol-2(3H)-yl]-8-methoxyquinolines with Potent Antibacterial Activity against Respiratory Pathogens. J Med Chem 2018; 61:7234-7244. [DOI: 10.1021/acs.jmedchem.8b00644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Takashi Odagiri
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hiroaki Inagaki
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masatoshi Nagamochi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takahiro Kitamura
- R&D Division, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Satoshi Komoriya
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hisashi Takahashi
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
23
|
Mao X, Hu Z, Wang Q, Zhang N, Zhou S, Peng Y, Zheng J. Nitidine Chloride Is a Mechanism-Based Inactivator of CYP2D6. Drug Metab Dispos 2018; 46:1137-1145. [PMID: 29773554 DOI: 10.1124/dmd.117.079780] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/11/2018] [Indexed: 12/11/2022] Open
Abstract
Nitidine chloride (NC) is a benzophenanthridine alkaloid isolated from the roots of Zanthoxylum nitidum (Roxb.) DC, a widely used traditional herbal medicine. Several reports have revealed NC's multiple pharmacologic properties. The inhibitory effects of NC on human cytochrome P450 enzymes were investigated in the present study. We found that NC caused time- and concentration-dependent inhibition of CYP2D6, and more than 50% of CYP2D6 activity was suppressed after a 15-minute incubation with NC at 100 μM in the primary incubation mixtures, with KI of 4.36 μM, kinact of 0.052 minute-1, and a partition ratio of approximately 290. Moreover, the loss of CYP2D6 activity required the presence of NADPH. Superoxide dismutase/catalase and glutathione showed minor protection against the NC-induced enzyme inhibition. Quinidine as a competitive inhibitor of CYP2D6 slowed down the inactivation by NC. Trapping experiments using N-acetylcysteine demonstrated that quinone and/or carbene intermediate(s) were/was generated in human liver microsomal incubations with NC. In addition, potassium ferricyanide prevented the enzyme from the inactivation mediated by NC, which provided evidence that inhibition of CYP2D6 resulted from heme destruction by the formation of a carbene-iron complex. CYP1A2 was found to be the major enzyme involved in the generation of NC quinone metabolites. In conclusion, NC is a mechanism-based inactivator of CYP2D6. The generation of a carbene intermediate might be mainly responsible for the enzyme inactivation.
Collapse
Affiliation(s)
- Xu Mao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China (X.M., Z.H., Q.W., N.Z., S.Z., Y.P., J.Z.); State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province and Guizhou Medical University, Guiyang, Guizhou, P. R. China (J.Z.)
| | - Zixia Hu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China (X.M., Z.H., Q.W., N.Z., S.Z., Y.P., J.Z.); State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province and Guizhou Medical University, Guiyang, Guizhou, P. R. China (J.Z.)
| | - Qian Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China (X.M., Z.H., Q.W., N.Z., S.Z., Y.P., J.Z.); State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province and Guizhou Medical University, Guiyang, Guizhou, P. R. China (J.Z.)
| | - Na Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China (X.M., Z.H., Q.W., N.Z., S.Z., Y.P., J.Z.); State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province and Guizhou Medical University, Guiyang, Guizhou, P. R. China (J.Z.)
| | - Shenzhi Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China (X.M., Z.H., Q.W., N.Z., S.Z., Y.P., J.Z.); State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province and Guizhou Medical University, Guiyang, Guizhou, P. R. China (J.Z.)
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China (X.M., Z.H., Q.W., N.Z., S.Z., Y.P., J.Z.); State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province and Guizhou Medical University, Guiyang, Guizhou, P. R. China (J.Z.)
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P. R. China (X.M., Z.H., Q.W., N.Z., S.Z., Y.P., J.Z.); State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province and Guizhou Medical University, Guiyang, Guizhou, P. R. China (J.Z.)
| |
Collapse
|
24
|
Yadav J, Korzekwa K, Nagar S. Improved Predictions of Drug-Drug Interactions Mediated by Time-Dependent Inhibition of CYP3A. Mol Pharm 2018; 15:1979-1995. [PMID: 29608318 PMCID: PMC5938745 DOI: 10.1021/acs.molpharmaceut.8b00129] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Time-dependent inactivation (TDI) of cytochrome P450s (CYPs) is a leading cause of clinical drug-drug interactions (DDIs). Current methods tend to overpredict DDIs. In this study, a numerical approach was used to model complex CYP3A TDI in human-liver microsomes. The inhibitors evaluated included troleandomycin (TAO), erythromycin (ERY), verapamil (VER), and diltiazem (DTZ) along with the primary metabolites N-demethyl erythromycin (NDE), norverapamil (NV), and N-desmethyl diltiazem (NDD). The complexities incorporated into the models included multiple-binding kinetics, quasi-irreversible inactivation, sequential metabolism, inhibitor depletion, and membrane partitioning. The resulting inactivation parameters were incorporated into static in vitro-in vivo correlation (IVIVC) models to predict clinical DDIs. For 77 clinically observed DDIs, with a hepatic-CYP3A-synthesis-rate constant of 0.000 146 min-1, the average fold difference between the observed and predicted DDIs was 3.17 for the standard replot method and 1.45 for the numerical method. Similar results were obtained using a synthesis-rate constant of 0.000 32 min-1. These results suggest that numerical methods can successfully model complex in vitro TDI kinetics and that the resulting DDI predictions are more accurate than those obtained with the standard replot approach.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Swati Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
25
|
Kosaka M, Kosugi Y, Hirabayashi H. Risk Assessment Using Cytochrome P450 Time-Dependent Inhibition Assays at Single Time and Concentration in the Early Stage of Drug Discovery. J Pharm Sci 2017; 106:2839-2846. [DOI: 10.1016/j.xphs.2017.04.077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/28/2017] [Accepted: 04/28/2017] [Indexed: 02/07/2023]
|
26
|
Inactivation kinetics and residual activity of CYP3A4 after treatment with erythromycin. Biopharm Drug Dispos 2017; 38:420-425. [DOI: 10.1002/bdd.2078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/19/2017] [Accepted: 04/11/2017] [Indexed: 11/07/2022]
|
27
|
Watanabe A, Takakusa H, Kimura T, Inoue SI, Kusuhara H, Ando O. Difference in Mechanism-Based Inhibition of Cytochrome P450 3A4 and 3A5 by a Series of Fluoroquinolone Antibacterial Agents. Drug Metab Dispos 2016; 45:336-341. [PMID: 27974381 DOI: 10.1124/dmd.116.073783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/13/2016] [Indexed: 11/22/2022] Open
Abstract
A series of fluoroquinolone antibacterial compounds were found to be irreversible (compounds 1-5) and quasi-irreversible (compounds 6-9) inhibitors of CYP3A4. The purpose of this study was to evaluate their mechanism-based inhibition (MBI) potency against CYP3A5. Compounds 1-5 were also irreversible inhibitors of CYP3A5, whereas compounds 6-9 showed neither irreversible nor quasi-irreversible inhibition of CYP3A5. Compounds 6 and 8 did not form a metabolite-intermediate complex with the heme of CYP3A5 during incubation. The structural analysis of the metabolites after incubation of compounds 1 and 6 with CYP3A5 revealed that their metabolites were identical to those produced by CYP3A4, including the precursors of which are speculated to account for the MBI of CYP3A4. The homology modeling of CYP3A5 suggests that four residues around the nitroso intermediate of compound 6 in the substrate-binding pocket of CYP3A4 correspond with the bulkier residues in CYP3A5-especially Phe210 in CYP3A5-which might contribute to the steric hindrance with the nitroso intermediate of compound 6. The substrate-binding pocket structure of CYP3A5 might prevent the nitroso intermediate from coordinate binding with the heme, thereby preventing quasi-irreversible inhibition. Our study may provide new insights into the observable differences between the inhibition of CYP3A4 and CYP3A5.
Collapse
Affiliation(s)
- Akiko Watanabe
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Company, Ltd., Tokyo, Japan (A.W., H.T., S.I., O.A.); Structural Biology Group, Biological Research Department, Daiichi Sankyo RD Novare Company, Ltd., Tokyo, Japan (T.K.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Hideo Takakusa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Company, Ltd., Tokyo, Japan (A.W., H.T., S.I., O.A.); Structural Biology Group, Biological Research Department, Daiichi Sankyo RD Novare Company, Ltd., Tokyo, Japan (T.K.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Takako Kimura
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Company, Ltd., Tokyo, Japan (A.W., H.T., S.I., O.A.); Structural Biology Group, Biological Research Department, Daiichi Sankyo RD Novare Company, Ltd., Tokyo, Japan (T.K.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Shin-Ichi Inoue
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Company, Ltd., Tokyo, Japan (A.W., H.T., S.I., O.A.); Structural Biology Group, Biological Research Department, Daiichi Sankyo RD Novare Company, Ltd., Tokyo, Japan (T.K.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Hiroyuki Kusuhara
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Company, Ltd., Tokyo, Japan (A.W., H.T., S.I., O.A.); Structural Biology Group, Biological Research Department, Daiichi Sankyo RD Novare Company, Ltd., Tokyo, Japan (T.K.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| | - Osamu Ando
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Company, Ltd., Tokyo, Japan (A.W., H.T., S.I., O.A.); Structural Biology Group, Biological Research Department, Daiichi Sankyo RD Novare Company, Ltd., Tokyo, Japan (T.K.); and Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.)
| |
Collapse
|
28
|
Watanabe A, Takakusa H, Kimura T, Inoue SI, Kusuhara H, Ando O. Analysis of Mechanism-Based Inhibition of CYP 3A4 by a Series of Fluoroquinolone Antibacterial Agents. Drug Metab Dispos 2016; 44:1608-16. [DOI: 10.1124/dmd.116.071654] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/27/2016] [Indexed: 11/22/2022] Open
|
29
|
Shibata Y, Kagechika K, Ota M, Yamaguchi M, Setoguchi M, Kubo H, Chiba K, Takano H, Akiyama C, Ono M, Nishi M, Usui H. Synthesis and Structure-Activity Relationships of 2-Aminoacetamide Derivatives as Peroxisome Proliferator-Activated Receptor α/γ Dual Agonists. Chem Pharm Bull (Tokyo) 2016; 63:591-602. [PMID: 26235167 DOI: 10.1248/cpb.c15-00221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We describe the design, syntheses, and structure-activity relationships of novel zwitterionic compounds as nonthiazolidinedion-based peroxisome proliferator-activated receptor (PPAR) α/γ dual agonists. In our previous report, we obtained compound 1 showing potent PPARα/γ dual agonistic activities, together with a sufficient glucose-lowering effect in db/db mice. However, this compound possessed an issue, i.e., the 1,3,4-oxadiazole ring was not stable in acidic conditions. Thus, we carried out further optimization to improve the stability while maintaining the other favorable profile features including potent PPARα/γ dual agonistic activity. We addressed the issue by changing the oxadiazole ring to a bioisostere amide group. These amide derivatives were stable in acidic conditions and decreased plasma glucose and plasma triglyceride levels significantly without marked weight gain.
Collapse
|
30
|
Wong SG, Lee M, Wong BK. Single concentration loss of activity assay provides an improved assessment of drug-drug interaction risk compared to IC50-shift. Xenobiotica 2016; 46:953-66. [PMID: 26956546 DOI: 10.3109/00498254.2016.1143139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
1. The utility of two abbreviated, higher-throughput assays [IC50-shift and the loss of activity (LOA) assay] to evaluate time-dependent inhibition (TDI) of 24 structurally related compounds was compared. 2. Good correlation (R(2) = 0.90) between % inhibition and kinact/KI suggested that the LOA assay has utility as an indicator of TDI potential. Weaker correlation was observed for the shifted IC50 (IC50(T = 30)) (R(2) = 0.61) and the fold-shift in IC50 (R(2) = 0.17). 3. Primary mechanism for poor correlation was depletion of active enzyme at concentrations > 1 μM leading to greater than predicted inhibition in the IC50-shift assay. 4. Previously reported strong correlations between IC50(T = 30) and kinact/KI were found to be dependent on potent TDI compounds with kinact/KI > 30; correlation was reduced for moderate inhibitors (kinact/KI < 30). LOA assay maintained good correlation even when strong TDI compounds were excluded. 5. LOA assay (% Inhibition at 30 min, 10 μM) was a good predictor of in vivo DDI (AUCr), providing a graded response with low potential for false negatives or positives. IC50-shift assay had bias for over-predicting in vivo DDI and was more likely to identify false positives.
Collapse
Affiliation(s)
- Simon G Wong
- a Department of Pharmacokinetics and Drug Metabolism , Amgen , South San Francisco , CA , USA
| | - Mey Lee
- a Department of Pharmacokinetics and Drug Metabolism , Amgen , South San Francisco , CA , USA
| | - Bradley K Wong
- a Department of Pharmacokinetics and Drug Metabolism , Amgen , South San Francisco , CA , USA
| |
Collapse
|
31
|
Hong Y, Chia YMF, Yeo RH, Venkatesan G, Koh SK, Chai CLL, Zhou L, Kojodjojo P, Chan ECY. Inactivation of Human Cytochrome P450 3A4 and 3A5 by Dronedarone and N-Desbutyl Dronedarone. Mol Pharmacol 2015; 89:1-13. [DOI: 10.1124/mol.115.100891] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022] Open
|
32
|
A cocktail approach for assessing the in vitro activity of human cytochrome P450s: An overview of current methodologies. J Pharm Biomed Anal 2014; 101:221-37. [DOI: 10.1016/j.jpba.2014.03.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/13/2014] [Indexed: 01/27/2023]
|
33
|
Phenotyping of CYP450 in human liver microsomes using the cocktail approach. Anal Bioanal Chem 2014; 406:4875-87. [DOI: 10.1007/s00216-014-7915-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/07/2014] [Accepted: 05/19/2014] [Indexed: 01/20/2023]
|
34
|
Kozakai K, Yamada Y, Oshikata M, Kawase T, Suzuki E, Haramaki Y, Taniguchi H. Cocktail-substrate approach-based high-throughput assay for evaluation of direct and time-dependent inhibition of multiple cytochrome P450 isoforms. Drug Metab Pharmacokinet 2013; 29:198-207. [PMID: 24172718 DOI: 10.2133/dmpk.dmpk-13-rg-093] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Avoiding drug-drug interactions (DDIs) mediated through inhibition of cytochrome P450 (CYP) activity is highly desirable. Direct inhibition (DI) of CYP through new chemical entities (NCEs) or time-dependent inhibition (TDI) through reactive metabolites should be elucidated at an early stage of drug discovery research. In particular, TDI of CYP occurring through reactive metabolites may be irreversible and even sustained, causing far more serious DDIs for TDIs than for DIs. Furthermore, it is important to ascertain whether an NCE inhibits multiple CYP isoforms. Hence, using a cocktail-substrate approach that we previously established (in which the activity of 8 CYP isoforms is simultaneously evaluated in a single run), we evaluated the IC50 values of direct inhibitors and TDI parameters (kobs, shifted IC50, KI and kinact) of time-dependent inhibitors that affect multiple CYP isoforms. The IC50 values for 8 CYP isoforms obtained using the cocktail-substrate approach were nearly identical to values previously reported. The TDI parameters for CYP1A2, 2C9, 2C19, 2D6, and CYP3A4/5 obtained using the cocktail-substrate approach were also nearly identical to those obtained using a single-substrate approach. Thus, the cocktail-substrate approach is useful for evaluating DI and TDI in the early stages of drug discovery and development processes.
Collapse
Affiliation(s)
- Kazumasa Kozakai
- Department of Regenerative Medicine, Yokohama City University School of Medicine
| | | | | | | | | | | | | |
Collapse
|
35
|
Brantley SJ, Graf TN, Oberlies NH, Paine MF. A systematic approach to evaluate herb-drug interaction mechanisms: investigation of milk thistle extracts and eight isolated constituents as CYP3A inhibitors. Drug Metab Dispos 2013; 41:1662-70. [PMID: 23801821 PMCID: PMC3876807 DOI: 10.1124/dmd.113.052563] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 06/25/2013] [Indexed: 10/26/2022] Open
Abstract
Despite increasing recognition of potential untoward interactions between herbal products and conventional medications, a standard system for prospective assessment of these interactions remains elusive. This information gap was addressed by evaluating the drug interaction liability of the model herbal product milk thistle (Silybum marianum) with the CYP3A probe substrate midazolam. The inhibitory effects of commercially available milk thistle extracts and isolated constituents on midazolam 1'-hydroxylation were screened using human liver and intestinal microsomes. Relative to vehicle, the extract silymarin and constituents silybin A, isosilybin A, isosilybin B, and silychristin at 100 μM demonstrated >50% inhibition of CYP3A activity with at least one microsomal preparation, prompting IC50 determination. The IC50s for isosilybin B and silychristin were ∼60 and 90 μM, respectively, whereas those for the remaining constituents were >100 μM. Extracts and constituents that contained the 1,4-dioxane moiety demonstrated a >1.5-fold shift in IC50 when tested as potential mechanism-based inhibitors. The semipurified extract, silibinin, and the two associated constituents (silybin A and silybin B) demonstrated mechanism-based inhibition of recombinant CYP3A4 (KI, ∼100 μM; kinact, ∼0.20 min(-1)) but not microsomal CYP3A activity. The maximum predicted increases in midazolam area under the curve using the static mechanistic equation and recombinant CYP3A4 data were 1.75-fold, which may necessitate clinical assessment. Evaluation of the interaction liability of single herbal product constituents, in addition to commercially available extracts, will enable elucidation of mechanisms underlying potential clinically significant herb-drug interactions. Application of this framework to other herbal products would permit predictions of herb-drug interactions and assist in prioritizing clinical evaluation.
Collapse
Affiliation(s)
- Scott J Brantley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | |
Collapse
|
36
|
Berry LM, Zhao Z, Lin MHJ. Dynamic modeling of cytochrome P450 inhibition in vitro: impact of inhibitor depletion on IC₅₀ shift. Drug Metab Dispos 2013; 41:1433-41. [PMID: 23649703 DOI: 10.1124/dmd.113.051508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The impact of inhibitor depletion on the determination of shifted IC₅₀ (IC₅₀ determined after 30 minutes of preincubation with inhibitor) is examined. In addition, IC₅₀-shift data are analyzed using a mechanistic model that incorporates the processes of inhibitor depletion, as well as reversible and time-dependent inhibition. Anomalies such as a smaller-than-expected shift in IC₅₀ and even increases in IC₅₀ with preincubation were explained by the depletion of inhibitor during the preincubation. The IC₅₀-shift assay remains a viable approach to characterizing a wide range of reversible and time-dependent inhibitors. However, as with more traditional time-dependent inactivation methods, it is recommended that IC₅₀-shift experimental data be interpreted with some knowledge of the magnitude of inhibitor depletion. For the most realistic classification of time-dependent inhibitors using IC₅₀-shift methods, shifted IC₅₀ should be calculated using observed inhibitor concentrations at the end of the incubation rather than nominal inhibitor concentrations. Finally, a mechanistic model that includes key processes, such as competitive inhibition, enzyme inactivation, and inhibitor depletion, can be used to describe accurately the observed IC₅₀ and shifted IC₅₀ curves. For compounds showing an IC₅₀ fold shift >1.5 based on the observed inhibitor concentrations, reanalyzing the IC₅₀-shift data using the mechanistic model appeared to allow for reasonable estimation of Ki, KI, and kinact directly from the IC₅₀ shift experiments.
Collapse
Affiliation(s)
- Loren M Berry
- Pharmacokinetics and Drug Metabolism, Amgen, Inc., 360 Binney St., Cambridge, MA 02142, USA.
| | | | | |
Collapse
|
37
|
Nakamura Y, Fujimoto T, Ogawa Y, Namiki H, Suzuki S, Asano M, Sugita C, Mochizuki A, Miyazaki S, Tamaki K, Nagai Y, Inoue SI, Nagayama T, Kato M, Chiba K, Takasuna K, Nishi T. Lead optimization of 5-amino-6-(2,2-dimethyl-5-oxo-4-phenylpiperazin-1-yl)-4-hydroxyhexanamides to reduce a cardiac safety issue: discovery of DS-8108b, an orally active renin inhibitor. Bioorg Med Chem 2013; 21:3175-96. [PMID: 23598247 DOI: 10.1016/j.bmc.2013.03.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 03/11/2013] [Accepted: 03/15/2013] [Indexed: 01/26/2023]
Abstract
With the aim to address an undesired cardiac issue observed with our related compound in the recently disclosed novel series of renin inhibitors, further chemical modifications of this series were performed. Extensive structure-activity relationships studies as well as in vivo cardiac studies using the electrophysiology rat model led to the discovery of clinical candidate trans-adamantan-1-ol analogue 56 (DS-8108b) as a potent renin inhibitor with reduced potential cardiac risk. Oral administration of single doses of 3 and 10 mg/kg of 56 in cynomolgus monkeys pre-treated with furosemide led to significant reduction of mean arterial blood pressure for more than 12 h.
Collapse
Affiliation(s)
- Yuji Nakamura
- Lead Discovery & Optimization Research Laboratories I, Daiichi Sankyo Co., Ltd, 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Imamura Y, Murayama N, Okudaira N, Kurihara A, Inoue K, Yuasa H, Izumi T, Kusuhara H, Sugiyama Y. Effect of the fluoroquinolone antibacterial agent DX-619 on the apparent formation and renal clearances of 6β-hydroxycortisol, an endogenous probe for CYP3A4 inhibition, in healthy subjects. Pharm Res 2012; 30:447-57. [PMID: 23073666 DOI: 10.1007/s11095-012-0890-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 09/13/2012] [Indexed: 01/20/2023]
Abstract
PURPOSE To examine the effect of the fluoroquinolone DX-619 on CYP3A4 and urinary excretion of 6β-hydroxycortisol, an endogenous probe of hepatic CYP3A4 activity, in healthy subjects. METHODS The effect of DX-619 on CYP3A4 was examined in human liver microsomes. The apparent formation and renal clearance of 6β-hydroxycortisol (CL(6β-OHF) and CL(renal,6β-OHF), respectively) were determined in placebo- and DX-619-treated subjects. 6β-hydroxycortisol uptake was determined in HEK293 cells expressing OAT1, OAT3, OCT2, MATE1, and MATE2-K. RESULTS DX-619 was a mechanism-based inhibitor of CYP3A4, with K(I) and k(inact) of 67.9 ± 7.3 μmol/l and 0.0730 ± 0.0033 min(-1), respectively. Pharmacokinetic simulation suggested in vivo relevance of CYP3A4 inhibition by DX-619. CL(6β-OHF) and CL(renal,6β-OHF) were decreased 72% and 70%, respectively, on day 15 in DX-619-treated group compared with placebo (P < 0.05). 6β-hydroxycortisol was a substrate of OAT3 (K(m) = 183 ± 25 μmol/l), OCT2, MATE1, and MATE2-K. Maximum unbound concentration of DX-619 (9.1 ± 0.4 μmol/l) was above K(i) of DX-619 for MATE1 (4.32 ± 0.79 μmol/l). CONCLUSIONS DX-619 caused a moderate inhibition of hepatic CYP3A4-mediated formation and significant inhibition of MATE-mediated efflux of 6β-hydroxycortisol into urine. Caution is needed in applying CL(6β-OHF) as an index of hepatic CYP3A4 activity without evaluating CL(renal,6β-OHF).
Collapse
Affiliation(s)
- Yuichiro Imamura
- Drug Metabolism & Pharmacokinetics Research Laboratories, R&D Division, Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Aasa J, Hu Y, Eklund G, Lindgren A, Baranczewski P, Malmquist J, Turek D, Bueters T. Effect of Solvents on the Time-Dependent Inhibition of CYP3A4 and the Biotransformation of AZD3839 in Human Liver Microsomes and Hepatocytes. Drug Metab Dispos 2012; 41:159-69. [DOI: 10.1124/dmd.112.047597] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
40
|
Assessment of drug–drug interactions caused by metabolism-dependent cytochrome P450 inhibition. Chem Biol Interact 2012; 198:49-56. [DOI: 10.1016/j.cbi.2012.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 05/15/2012] [Accepted: 05/21/2012] [Indexed: 01/25/2023]
|
41
|
Nakayama S, Takakusa H, Watanabe A, Miyaji Y, Suzuki W, Sugiyama D, Shiosakai K, Honda K, Okudaira N, Izumi T, Okazaki O. Combination of GSH Trapping and Time-Dependent Inhibition Assays as a Predictive Method of Drugs Generating Highly Reactive Metabolites. Drug Metab Dispos 2011; 39:1247-54. [DOI: 10.1124/dmd.111.039180] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
42
|
Zimmerlin A, Trunzer M, Faller B. CYP3A Time-Dependent Inhibition Risk Assessment Validated with 400 Reference Drugs. Drug Metab Dispos 2011; 39:1039-46. [DOI: 10.1124/dmd.110.037911] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
43
|
Takakusa H, Wahlin MD, Zhao C, Hanson KL, New LS, Chan ECY, Nelson SD. Metabolic intermediate complex formation of human cytochrome P450 3A4 by lapatinib. Drug Metab Dispos 2011; 39:1022-30. [PMID: 21363997 DOI: 10.1124/dmd.110.037531] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Lapatinib, an oral breast cancer drug, has recently been reported to be a mechanism-based inactivator of cytochrome P450 (P450) 3A4 and also an idiosyncratic hepatotoxicant. It was suggested that formation of a reactive quinoneimine metabolite was involved in mechanism-based inactivation (MBI) and/or hepatotoxicity. We investigated the mechanism of MBI of P450 3A4 by lapatinib. Liquid chromatography-mass spectrometry analysis of P450 3A4 after incubation with lapatinib did not show any peak corresponding to irreversible modifications. The enzymatic activity inactivated by lapatinib was completely restored by the addition of potassium ferricyanide. These results indicate that the mechanism of MBI by lapatinib is quasi-irreversible and mediated via metabolic intermediate complex (MI complex) formation. This finding was verified by the increase in a signature Soret absorbance at approximately 455 nm. Two amine oxidation products of the metabolism of lapatinib by P450 3A4 were characterized: N-hydroxy lapatinib (M3) and the oxime form of N-dealkylated lapatinib (M2), suggesting that a nitroso or another related intermediate generated from M3 is involved in MI complex formation. In contrast, P450 3A5 was much less susceptible to MBI by lapatinib via MI complex formation than P450 3A4. In addition, P450 3A5 had a significantly lower ability than 3A4 to generate M3, consistent with N-hydroxylation as the initial step in the pathway to MI complex formation. In conclusion, our results demonstrate that the primary mechanism for MBI of P450 3A4 by lapatinib is not irreversible modification by the quinoneimine metabolite, but quasi-irreversible MI complex formation mediated via oxidation of the secondary amine group of lapatinib.
Collapse
Affiliation(s)
- Hideo Takakusa
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Nishiya Y, Nakamura K, Okudaira N, Abe K, Kobayashi N, Okazaki O. Effects of organic solvents on the time-dependent inhibition of CYP3A4 by diazepam. Xenobiotica 2009; 40:1-8. [DOI: 10.3109/00498250903337392] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
45
|
The P450 oxidoreductase genotype is associated with CYP3A activity in vivo as measured by the midazolam phenotyping test. Pharmacogenet Genomics 2009; 19:877-83. [DOI: 10.1097/fpc.0b013e32833225e7] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
46
|
Mori K, Hashimoto H, Takatsu H, Tsuda-Tsukimoto M, Kume T. Cocktail-substrate assay system for mechanism-based inhibition of CYP2C9, CYP2D6, and CYP3A using human liver microsomes at an early stage of drug development. Xenobiotica 2009; 39:415-22. [PMID: 19480547 DOI: 10.1080/00498250902822204] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We established a mechanism-based inhibition cocktail-substrate assay system using human liver microsomes and drug-probe substrates that enabled simultaneous estimation of the inactivation of main cytochrome P450 (CYP) enzymes, CYP2C9, CYP2D6, and CYP3A, in drug metabolism. The inactivation kinetic parameters of typical mechanism-based inhibitors, tienilic acid, paroxetine, and erythromycin, for each enzyme in the cocktail-substrate assay were almost in agreement with the values obtained in the single-substrate assay. Using this system, we confirmed that multiple CYP inactivation caused by mechanism-based inhibitors such as isoniazid and amiodarone could be detected simultaneously. Mechanism-based inhibition potency can be estimated by the determination of the observed inactivation rate constants (k(obs)) at a single concentration of test compounds because the k(obs) of eleven CYP3A inactivators at 10 microM in the assay system nearly corresponded to k(inact)/K(I) values, an indicator of a compound's propensity to alter the activity of a CYP in vivo (R(2) = 0.97). Therefore, this cocktail-substrate assay is considered to be a powerful tool for evaluating mechanism-based inhibition at an early stage of drug development.
Collapse
Affiliation(s)
- K Mori
- Drug Metabolism and Pharmacokinetic Research Laboratory, Mitsubishi Tanabe Pharma Corporation, Toda, Japan
| | | | | | | | | |
Collapse
|
47
|
Ogasawara A, Negishi I, Kozakai K, Kume T. In Vivo Evaluation of Drug-Drug Interaction via Mechanism-Based Inhibition by Macrolide Antibiotics in Cynomolgus Monkeys. Drug Metab Dispos 2009; 37:2127-36. [DOI: 10.1124/dmd.109.028969] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
48
|
Grimm SW, Einolf HJ, Hall SD, He K, Lim HK, Ling KHJ, Lu C, Nomeir AA, Seibert E, Skordos KW, Tonn GR, Van Horn R, Wang RW, Wong YN, Yang TJ, Obach RS. The Conduct of in Vitro Studies to Address Time-Dependent Inhibition of Drug-Metabolizing Enzymes: A Perspective of the Pharmaceutical Research and Manufacturers of America. Drug Metab Dispos 2009; 37:1355-70. [DOI: 10.1124/dmd.109.026716] [Citation(s) in RCA: 239] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
49
|
Sekiguchi N, Kato M, Takada M, Watanabe H, Higashida A, Sakai S, Ishigai M, Aso Y. In vivoapproach for the evaluation of mechanism-based inhibition of cytochrome P450 3A in rats. Xenobiotica 2008; 38:368-81. [DOI: 10.1080/00498250701851891] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
50
|
Bui PH, Quesada A, Handforth A, Hankinson O. The mibefradil derivative NNC55-0396, a specific T-type calcium channel antagonist, exhibits less CYP3A4 inhibition than mibefradil. Drug Metab Dispos 2008; 36:1291-9. [PMID: 18411403 DOI: 10.1124/dmd.107.020115] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A novel mibefradil derivative, NNC55-0396, designed to be hydrolysis-resistant, was shown to be a selective T-type Ca(2+) channel inhibitor without L-type Ca(2+) channel efficacy. However, its effects on cytochromes P450 (P450s) have not previously been examined. We investigated the inhibitory effects of NNC55-0396 toward seven major recombinant human P450s--CYP3A4, CYP2D6, CYP1A2, CYP2C9, CYP2C8, CYPC19, and CYP2E1--and compared its effects with those of mibefradil and its hydrolyzed metabolite, Ro40-5966. Our results show that CYP3A4 and CYP2D6 are the two P450s most affected by mibefradil, Ro40-5966, and NNC55-0396. Mibefradil (IC(50) = 33 +/- 3 nM, K(i) = 23 +/- 0.5 nM) and Ro40-5966 (IC(50) = 30 +/- 7.8 nM, K(i) = 21 +/- 2.8 nM) have a 9- to 10-fold greater inhibitory activity toward recombinant CYP3A4 benzyloxy-4-trifluoromethylcoumarin-O-debenzylation activity than NNC55-0396 (IC(50) = 300 +/- 30 nM, K(i) = 210 +/- 6 nM). More dramatically, mibefradil (IC(50) = 566 +/- 71 nM, K(i) = 202 +/- 39 nM) shows 19-fold higher inhibition of CYP3A-associated testosterone 6beta-hydroxylase activity in human liver microsomes compared with NNC55-0396 (IC(50) = 11 +/- 1.1 microM, K(i) = 3.9 +/- 0.4 microM). Loss of testosterone 6beta-hydroxylase activity by recombinant CYP3A4 was shown to be time- and concentration-dependent with both compounds. However, NNC55-0396 (K(I) = 3.87 microM, K(inact) = 0.061/min) is a much less potent mechanism-based inhibitor than mibefradil (K(I) = 83 nM, K(inact) = 0.048/min). In contrast, NNC55-0396 (IC(50) = 29 +/- 1.2 nM, K(i) = 2.8 +/- 0.3 nM) and Ro40-5966 (IC(50) = 46 +/- 11 nM, K(i) = 4.5 +/- 0.02 nM) have a 3- to 4-fold greater inhibitory activity toward recombinant CYP2D6 than mibefradil (IC(50) = 129 +/- 21 nM, K(i) = 12.7 +/- 0.9 nM). Our results suggest that NNC55-0396 could be a more favorable T-type Ca(2+) antagonist than its parent compound, mibefradil, which was withdrawn from the market because of strong inhibition of CYP3A4.
Collapse
Affiliation(s)
- Peter H Bui
- Departmental of Pathology and Laboratory Medicine, University of California at Los Angeles, 650 Charles Young Drive, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|