1
|
Ning J, Tian Z, Wang J, Yan F, Shi C, Zhang S, Feng L, Shu X, Cui J, James TD, Ma X. Rational Molecular Design of a Fluorescent Probe for Selectively Sensing Human Cytochrome P450 2D6. Angew Chem Int Ed Engl 2024; 63:e202409217. [PMID: 38989537 DOI: 10.1002/anie.202409217] [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: 05/15/2024] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 07/12/2024]
Abstract
Cytochrome P450 2D6 (CYP2D6) is a key enzyme that mediates the metabolism of various drugs and endogenous substances in humans. However, its biological role in drug-drug interactions especially mechanism-based inactivation (MBI), and various diseases remains poorly understood, owing to the lack of molecular tools suitable for selectively monitoring CYP2D6 in complex biological systems. Herein, using a tailored molecular strategy, we developed a fluorescent probe BDPM for CYP2D6. BDPM exhibits excellent specificity and imaging capability for CYP2D6, making it suitable for the real-time monitoring of endogenous CYP2D6 activity in living bio-samples. Therefore, our tailored strategy proved useful for constructing the highly selective and enzyme-activated fluorescent probes. BDPM as a molecular tool to explore the critical roles of CYP2D6 in the pathogenesis of diseases, high-throughput screening of inhibitors and intensive investigation of CYP2D6-induced MBI in natural systems.
Collapse
Affiliation(s)
- Jing Ning
- The 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
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- Beijing DP Technology Co., Ltd., Beijing, 100080, China
| | - Fei Yan
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Chao Shi
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Shujing Zhang
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Lei Feng
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China
| | - Xiaohong Shu
- College of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian, 116044, 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
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| |
Collapse
|
2
|
Tan BH, Ahemad N, Pan Y, Ong CE. Mechanism-based inactivation of cytochromes P450: implications in drug interactions and pharmacotherapy. Xenobiotica 2024; 54:575-598. [PMID: 39175333 DOI: 10.1080/00498254.2024.2395557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Cytochrome P40 (CYP) enzymes dominate the metabolism of numerous endogenous and xenobiotic substances. While it is commonly believed that CYP-catalysed reactions result in the detoxication of foreign substances, these reactions can also yield reactive intermediates that can bind to cellular macromolecules to cause cytotoxicity or irreversibly inactivate CYPs that create them.Mechanism-based inactivation (MBI) produces either irreversible or quasi-irreversible inactivation and is commonly caused by CYP metabolic bioactivation to an electrophilic reactive intermediate. Many drugs that have been known to cause MBI in CYPs have been discovered as perpetrators in drug-drug interactions throughout the last 20-30 years.This review will highlight the key findings from the recent literature about the mechanisms of CYP enzyme inhibition, with a focus on the broad mechanistic elements of MBI for widely used drugs linked to the phenomenon. There will also be a brief discussion of the clinical or pharmacokinetic consequences of CYP inactivation with regard to drug interaction and toxicity risk.Gaining knowledge about the selective inactivation of CYPs by common therapeutic drugs helps with the assessment of factors that affect the systemic clearance of co-administered drugs and improves comprehension of anticipated interactions with other drugs or xenobiotics.
Collapse
Affiliation(s)
- Boon Hooi Tan
- Division of Applied Biomedical Sciences and Biotechnology, International Medical University, Kuala Lumpur, Malaysia
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Selangor, Malaysia
| | - Yan Pan
- Department of Biomedical Science, University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Chin Eng Ong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
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
|
4
|
Yamaguchi Y, Nishizono N, Oda K. Evaluation of Synthesized Ester or Amide Coumarin Derivatives on Aromatase Inhibitory Activity. Biol Pharm Bull 2020; 43:1179-1187. [DOI: 10.1248/bpb.b20-00035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuki Yamaguchi
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Naozumi Nishizono
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Kazuaki Oda
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| |
Collapse
|
5
|
Yong YF, Tan SC, Liew MWO, Yaacob NS. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) method development for screening of potential tamoxifen-drug/herb interaction via in vitro cytochrome P450 inhibition assay. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1148:122148. [PMID: 32416571 DOI: 10.1016/j.jchromb.2020.122148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/19/2020] [Accepted: 05/04/2020] [Indexed: 11/30/2022]
Abstract
Screening for potential drug-drug interaction (DDI) or herb-drug interaction (HDI) using in vitro cytochrome P450 inhibition (IVCI) assays requires robust analytical methods with high sensitivity and reproducibility. Utilization of liquid chromatography-mass spectrometry (LC-MS) for analyte quantification is often hampered by the presence of non-volatile IVCI sample buffer constituents that often results in ion suppression. In this study, to enable screening of drug interactions involving tamoxifen (TAM) metabolism using IVCI-LC-MS/MS, a liquid-liquid extraction (LLE) method was developed and optimized for sample clean-up. Utilization of chloroform as extraction solvent and adjustment of sample pH to 11 was found to result in satisfactory recovery (>70%) and low ion suppression (<19%). A LC-MS/MS method was subsequently developed and validated for simultaneous quantification of major TAM metabolites, such as N-desmethyltamoxifen (NDT), endoxifen (EDF) and 4-hydroxytamoxifen (HTF) to enable IVCI sample analysis. Satisfactory separation of E-/Z-isomers of endoxifen with peak resolution (Rs) of 1.9 was achieved. Accuracy and precision of the method was verified within the linear range of 0-50 ng/mL for NDT, 0-25 ng/mL for HTF and 0-25 ng/mL for EDF (E/Z isomers). Inhibitory potency (IC50, Ki and mode of inhibition) of known CYP inhibitors and Strobilanthes crispus extract was then evaluated using the validated method. In summary, the results demonstrated applicability of the developed LLE and validated LC-MS/MS method for in vitro screening of DDI and HDI involving TAM metabolism.
Collapse
Affiliation(s)
- Y F Yong
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - S C Tan
- Usains Biomics Laboratory Testing Services Sdn. Bhd., Universiti Sains Malaysia, Minden 11800, Penang, Malaysia
| | - Mervyn W O Liew
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - N S Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
6
|
Tolledo EC, Miksys S, Gonzalez FJ, Tyndale RF. Propranolol is a mechanism-based inhibitor of CYP2D and CYP2D6 in humanized CYP2D6-transgenic mice: Effects on activity and drug responses. Br J Pharmacol 2020; 177:701-712. [PMID: 31648367 PMCID: PMC7012948 DOI: 10.1111/bph.14884] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/16/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Genetics and drug interactions contribute to large interindividual variation in human CYP2D6 activity. Here, we have characterized propranolol inhibition of human and mouse CYP2D using transgenic (TG) mice, which express both mouse CYP2D and human CYP2D6, and wild-type (WT) mice. Our purpose was to develop a method for in vivo manipulation of CYP2D6 enzyme activity which could be used to investigate the role of CYP2D6 in drug-induced behaviours. EXPERIMENTAL APPROACH Dextromethorphan metabolism to dextrorphan was used to measure CYP2D activity and to characterize propranolol inhibition in vitro and in vivo. Effects of propranolol pretreatment (24 hr) on serum levels of the CYP2D6 substrate haloperidol and haloperidol-induced catalepsy were also studied. KEY RESULTS Dextrorphan formation velocity in vitro was threefold higher in liver microsomes of TG compared to WT mice. Propranolol acted as a mechanism-based inhibitor (MBI), inactivating CYP2D in liver microsomes from TG and WT mice, and humans. Pretreatment (24 hr) of TG and WT mice with 20 mg·kg-1 intraperitoneal propranolol reduced dextrorphan formation in vivo and by liver microsomes in vitro. Serum haloperidol levels and catalepsy were increased. CONCLUSIONS AND IMPLICATIONS Propranolol was a potent MBI of dextrorphan formation in liver microsomes from TG and WT mice, and humans. The inhibition parameters in TG overlapped with those in WT mice and in humans. Inhibition of CYP2D with propranolol in vivo in TG and WT mice altered drug responses, allowing further investigation of variations in CYP2D6 on drug interactions and drug responses.
Collapse
Affiliation(s)
- Edgor Cole Tolledo
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Department of Pharmacology & Toxicology, Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Sharon Miksys
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Department of Pharmacology & Toxicology, Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Rachel F. Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Department of Pharmacology & Toxicology, Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
7
|
Kahma H, Filppula AM, Launiainen T, Viinamäki J, Neuvonen M, Evangelista EA, Totah RA, Backman JT. Critical Differences between Enzyme Sources in Sensitivity to Detect Time-Dependent Inactivation of CYP2C8. Drug Metab Dispos 2019; 47:436-443. [PMID: 30709838 PMCID: PMC11022894 DOI: 10.1124/dmd.118.085498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Clopidogrel acyl-β-d-glucuronide is a mechanism-based inhibitor of cytochrome P450 2C8 in human liver microsomes (HLMs). However, time-dependent inactivation (TDI) of CYP2C8 could not be detected in an earlier study in human recombinant CYP2C8 (Supersomes). Here, we investigate whether different enzyme sources exhibit differences in detection of CYP2C8 TDI under identical experimental conditions. Inactivation of CYP2C8 by amiodarone (100 μM), clopidogrel acyl-β-d-glucuronide (100 μM), gemfibrozil 1-O-β-glucuronide (100 μM), and phenelzine (100 μM) was investigated in HLMs and three recombinant human CYP2C8 preparations (Supersomes, Bactosomes, and EasyCYP Bactosomes) using amodiaquine N-deethylation as the marker reaction. Furthermore, the inactivation kinetics of CYP2C8 by clopidogrel glucuronide (5-250 μM) was determined in Supersomes and Bactosomes. Amiodarone caused weak TDI in all enzyme preparations tested, while the extent of inactivation by clopidogrel glucuronide, gemfibrozil glucuronide, and phenelzine varied markedly between preparations, and even different Supersome lots. Both glucuronides caused strong inactivation of CYP2C8 in HLMs, Bactosomes and in one Supersome lot (>50% inhibition), but significant inactivation could not be reliably detected in other Supersome lots or EasyCYP Bactosomes. In Bactosomes, the concentration producing half of kinact (KI) and maximal inactivation rate (kinact) of clopidogrel glucuronide (14 μM and 0.054 minute-1) were similar to those determined previously in HLMs. Phenelzine caused strong inactivation of CYP2C8 in one Supersome lot (91% inhibition) but not in HLMs or other recombinant CYP2C8 preparations. In conclusion, different enzyme sources and different lots of the same recombinant enzyme preparation are not equally sensitive to detect inactivation of CYP2C8, suggesting that recombinant CYPs should be avoided when identifying mechanism-based inhibitors.
Collapse
Affiliation(s)
- Helinä Kahma
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Anne M Filppula
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Terhi Launiainen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Jenni Viinamäki
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Mikko Neuvonen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Eric A Evangelista
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Rheem A Totah
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Janne T Backman
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| |
Collapse
|
8
|
Lee E, Kim JH, Shon JC, Wu Z, Kim HJ, Gim M, Lee T, Liu KH. Terfenadone is a strong inhibitor of CYP2J2 present in the human liver and intestinal microsomes. Drug Metab Pharmacokinet 2018; 33:159-163. [DOI: 10.1016/j.dmpk.2018.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 11/07/2017] [Accepted: 02/23/2018] [Indexed: 10/17/2022]
|
9
|
Reinen J, Smit M, Wenker M. Evaluation of Strategies for the Assessment of Drug–Drug Interactions Involving Cytochrome P450 Enzymes. Eur J Drug Metab Pharmacokinet 2018; 43:737-750. [DOI: 10.1007/s13318-018-0485-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Burt HJ, Neuhoff S, Almond L, Gaohua L, Harwood MD, Jamei M, Rostami-Hodjegan A, Tucker GT, Rowland-Yeo K. Metformin and cimetidine: Physiologically based pharmacokinetic modelling to investigate transporter mediated drug-drug interactions. Eur J Pharm Sci 2016; 88:70-82. [PMID: 27019345 DOI: 10.1016/j.ejps.2016.03.020] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/10/2016] [Accepted: 03/22/2016] [Indexed: 01/11/2023]
Abstract
Metformin is used as a probe for OCT2 mediated transport when investigating possible DDIs with new chemical entities. The aim of the current study was to investigate the ability of physiologically-based pharmacokinetic (PBPK) models to simulate the effects of OCT and MATE inhibition by cimetidine on metformin kinetics. PBPK models were developed, incorporating mechanistic kidney and liver sub-models for metformin (OCT and MATE substrate) and a mechanistic kidney sub-model for cimetidine. The models were used to simulate inhibition of the MATE1, MATE2-K, OCT1 and OCT2 mediated transport of metformin by cimetidine. Assuming competitive inhibition and using cimetidine Ki values determined in vitro, the predicted metformin AUC ratio was 1.0 compared to an observed value of 1.46. The observed AUC ratio could only be recovered with this model when the cimetidine Ki for OCT2 was decreased 1000-fold or the Ki's for both OCT1 and OCT2 were decreased 500-fold. An alternative description of metformin renal transport by OCT1 and OCT2, incorporating electrochemical modulation of the rate of metformin uptake together with 8-18-fold decreases in cimetidine Ki's for OCTs and MATEs, allowed recovery of the extent of the observed effect of cimetidine on metformin AUC. While the final PBPK model has limitations, it demonstrates the benefit of allowing for the complexities of passive permeability combined with active cellular uptake modulated by an electrochemical gradient and active efflux.
Collapse
Affiliation(s)
- H J Burt
- Simcyp (a Certara Company), Sheffield, UK.
| | - S Neuhoff
- Simcyp (a Certara Company), Sheffield, UK.
| | - L Almond
- Simcyp (a Certara Company), Sheffield, UK.
| | - L Gaohua
- Simcyp (a Certara Company), Sheffield, UK.
| | | | - M Jamei
- Simcyp (a Certara Company), Sheffield, UK.
| | - A Rostami-Hodjegan
- Simcyp (a Certara Company), Sheffield, UK; Manchester Pharmacy School, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK.
| | - G T Tucker
- Medicine and Biomedical Sciences (emeritus), University of Sheffield, Sheffield, UK.
| | | |
Collapse
|
11
|
Akiyoshi T, Ishiuchi M, Imaoka A, Ohtani H. Variation in the inhibitory potency of terbinafine among genetic variants of CYP2D6. Drug Metab Pharmacokinet 2015. [PMID: 26195224 DOI: 10.1016/j.dmpk.2015.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cytochrome P450 2D6 (CYP2D6) is a highly polymorphic enzyme that is involved in the metabolism of many drugs. Terbinafine (TER) is a CYP2D6 inhibitor and causes persistent drug interactions in the clinical setting; however, its inhibitory mechanism and the differences in its inhibitory potency among genetic variants of CYP2D6 remain to be investigated. This study aimed to investigate the inhibitory mechanism of TER and the differences in its inhibitory potency among three CYP2D6 variants, CYP2D6.1, CYP2D6.2, and CYP2D6.10. In a competitive inhibition study, the metabolic activity of the CYP2D6 was assessed based on their demethylation of dextromethorphan in the presence or absence of TER, and the time-dependency of the inhibitory effects were examined by preincubating the enzymes with TER. TER had weaker inhibitory effects on CYP2D6.2 and CYP2D6.10 than on CYP2D6.1; i.e., TER exhibited Ki values (the concentration of inhibitor that results in half-maximal inhibition) of 0.0525, 0.355, and 1.85 μM for CYP2D6.1, CYP2D6.2, and CYP2D6.10, respectively. The inhibitory effects of TER were not time-dependent. Since TER's Ki value for CYP2D6.10 was 35.2-fold higher than its Ki value for CYP2D6.1, the CYP2D6 genotype of subjects should be taken into account when estimating the severity of drug interactions involving TER.
Collapse
Affiliation(s)
| | | | - Ayuko Imaoka
- Keio University, Faculty of Pharmacy, Tokyo, Japan
| | | |
Collapse
|
12
|
Sun J, Peng Y, Wu H, Zhang X, Zhong Y, Xiao Y, Zhang F, Qi H, Shang L, Zhu J, Sun Y, Liu K, Liu J, A J, Ho RJY, Wang G. Guanfu base A, an antiarrhythmic alkaloid of Aconitum coreanum, Is a CYP2D6 inhibitor of human, monkey, and dog isoforms. Drug Metab Dispos 2015; 43:713-24. [PMID: 25681130 DOI: 10.1124/dmd.114.060905] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Guanfu base A (GFA) is a novel heterocyclic antiarrhythmic drug isolated from Aconitum coreanum (Lèvl.) rapaics and is currently in a phase IV clinical trial in China. However, no study has investigated the influence of GFA on cytochrome P450 (P450) drug metabolism. We characterized the potency and specificity of GFA CYP2D inhibition based on dextromethorphan O-demethylation, a CYP2D6 probe substrate of activity in human, mouse, rat, dog, and monkey liver microsomes. In addition, (+)-bufuralol 1'-hydroxylation was used as a CYP2D6 probe for the recombinant form (rCYP2D6), 2D1 (rCYP2D1), and 2D2 (rCYP2D2) activities. Results show that GFA is a potent noncompetitive inhibitor of CYP2D6, with inhibition constant Ki = 1.20 ± 0.33 μM in human liver microsomes (HLMs) and Ki = 0.37 ± 0.16 μM for the human recombinant form (rCYP2D6). GFA is also a potent competitive inhibitor of CYP2D in monkey (Ki = 0.38 ± 0.12 μM) and dog (Ki = 2.4 ± 1.3 μM) microsomes. However, GFA has no inhibitory activity on mouse or rat CYP2Ds. GFA did not exhibit any inhibition activity on human recombinant CYP1A2, 2A6, 2C8, 2C19, 3A4, or 3A5, but showed slight inhibition of 2B6 and 2E1. Preincubation of HLMs and rCYP2D6 resulted in the inactivation of the enzyme, which was attenuated by GFA or quinidine. Beagle dogs treated intravenously with dextromethorphan (2 mg/ml) after pretreatment with GFA injection showed reduced CYP2D metabolic activity, with the Cmax of dextrorphan being one-third that of the saline-treated group and area under the plasma concentration-time curve half that of the saline-treated group. This study suggests that GFA is a specific CYP2D6 inhibitor that might play a role in CYP2D6 medicated drug-drug interaction.
Collapse
Affiliation(s)
- Jianguo Sun
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Ying Peng
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Hui Wu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Xueyuan Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Yunxi Zhong
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Yanan Xiao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Fengyi Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Huanhuan Qi
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Lili Shang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Jianping Zhu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Yue Sun
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Ke Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Jinghan Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Jiye A
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Rodney J Y Ho
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (J.S., Y.P., H.W., X.Z., Y.Z., Y.X., F.Z., H.Q., L.S., J.Z., Y.S., K.L., J.A., G.W.), and Department of Natural Medicinal Chemistry (J.L.), China Pharmaceutical University, Nanjing, China; and Department of Pharmaceutics, University of Washington, Seattle, Washington (R.J.Y.H.)
| |
Collapse
|
13
|
In vitro Inhibition of Methadone and Oxycodone Cytochrome P450-Dependent Metabolism: Reversible Inhibition by H2-Receptor Agonists and Proton-Pump Inhibitors. J Anal Toxicol 2013; 37:476-85. [DOI: 10.1093/jat/bkt060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
14
|
Martinez MN, Antonovic L, Court M, Dacasto M, Fink-Gremmels J, Kukanich B, Locuson C, Mealey K, Myers MJ, Trepanier L. Challenges in exploring the cytochrome P450 system as a source of variation in canine drug pharmacokinetics. Drug Metab Rev 2013; 45:218-30. [DOI: 10.3109/03602532.2013.765445] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
15
|
Fujioka Y, Kunze KL, Isoherranen N. Risk assessment of mechanism-based inactivation in drug-drug interactions. Drug Metab Dispos 2012; 40:1653-7. [PMID: 22685217 DOI: 10.1124/dmd.112.046649] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Drug-drug interactions (DDIs) that occur via mechanism-based inactivation of cytochrome P450 are of serious concern. Although several predictive models have been published, early risk assessment of MBIs is still challenging. For reversible inhibitors, the DDI risk categorization using [I]/K(i) ([I], the inhibitor concentration; K(i), the inhibition constant) is widely used in drug discovery and development. Although a simple and reliable methodology such as [I]/K(i) categorization for reversible inhibitors would be useful for mechanism-based inhibitors (MBIs), comprehensive analysis of an analogous measure reflecting in vitro potency for inactivation has not been reported. The aim of this study was to evaluate whether the term λ/k(deg) (λ, first-order inactivation rate at a given MBI concentration; k(deg), enzyme degradation rate constant) would be useful in the prediction of the in vivo DDI risk of MBIs. Twenty-one MBIs with both in vivo area under the curve (AUC) change of marker substrates and in vitro inactivation parameters were identified in the literature and analyzed. The results of this analysis show that in vivo DDIs with >2-fold change of object drug AUC can be identified with the cutoff value of λ/k(deg) = 1, where unbound steady-state C(max) is used for inhibitor concentration. However, the use of total C(max) led to great overprediction of DDI risk. The risk assessment using λ/k(deg) coupled with unbound C(max) can be useful for the DDI risk evaluation of MBIs in drug discovery and development.
Collapse
Affiliation(s)
- Yasushi Fujioka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
16
|
Lutz JD, Isoherranen N. In vitro-to-in vivo predictions of drug-drug interactions involving multiple reversible inhibitors. Expert Opin Drug Metab Toxicol 2012; 8:449-66. [PMID: 22384784 DOI: 10.1517/17425255.2012.667801] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Predictions of drug-drug interactions (DDIs) are commonly performed for single inhibitors, but interactions involving multiple inhibitors also frequently occur. Predictions of such interactions involving stereoisomer pairs, parent/metabolite combinations and simultaneously administered multiple inhibitors are increasing in importance. This review provides the framework for predicting inhibitory DDIs of multiple inhibitors with any combination of reversible inhibition mechanism. AREAS COVERED The review provides an overview of the reliability of the in vitro determined reversible inhibition mechanism. Furthermore, the article provides a method to predict DDIs for multiple reversible inhibitors that allows substituting the inhibition constant (K(i)) with an inhibitor affinity (IC(50)) value determined at S << K(M). EXPERT OPINION A better understanding and the prediction methods of DDIs, resulting from multiple inhibitors, are important. The inhibition mechanism of a reversible inhibitor is often equivocal across studies and unreliable. Determination of the K(i) requires the assignment of reversible inhibition mechanism but in vitro-to-in vivo prediction of DDI risk can be achieved for multiple inhibitors from estimates of the inhibitor affinity (IC(50)) only, regardless of the inhibition mechanism.
Collapse
Affiliation(s)
- Justin D Lutz
- University of Washington School of Pharmacy, Department of Pharmaceutics, Seattle, WA, USA
| | | |
Collapse
|
17
|
Yamaori S, Okamoto Y, Yamamoto I, Watanabe K. Cannabidiol, a Major Phytocannabinoid, As a Potent Atypical Inhibitor for CYP2D6. Drug Metab Dispos 2011; 39:2049-56. [DOI: 10.1124/dmd.111.041384] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
18
|
|
19
|
Sideras K, Ingle JN, Ames MM, Loprinzi CL, Mrazek DP, Black JL, Weinshilboum RM, Hawse JR, Spelsberg TC, Goetz MP. Coprescription of tamoxifen and medications that inhibit CYP2D6. J Clin Oncol 2010; 28:2768-76. [PMID: 20439629 PMCID: PMC2881853 DOI: 10.1200/jco.2009.23.8931] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 11/18/2009] [Indexed: 12/14/2022] Open
Abstract
Evidence has emerged that the clinical benefit of tamoxifen is related to the functional status of the hepatic metabolizing enzyme cytochrome P450 2D6 (CYP2D6). CYP2D6 is the key enzyme responsible for the generation of the potent tamoxifen metabolite, endoxifen. Multiple studies have examined the relationship of CYP2D6 status to breast cancer outcomes in tamoxifen-treated women; the majority of studies demonstrated that women with impaired CYP2D6 metabolism have lower endoxifen concentrations and a greater risk of breast cancer recurrence. As a result, practitioners must be aware that some of the most commonly prescribed medications coadministered with tamoxifen interfere with CYP2D6 function, thereby reducing endoxifen concentrations and potentially increasing the risk of breast cancer recurrence. After reviewing the published data regarding tamoxifen metabolism and the evidence relating CYP2D6 status to breast cancer outcomes in tamoxifen-treated patients, we are providing a guide for the use of medications that inhibit CYP2D6 in patients administered tamoxifen.
Collapse
Affiliation(s)
- Kostandinos Sideras
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - James N. Ingle
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Matthew M. Ames
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Charles L. Loprinzi
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - David P. Mrazek
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - John L. Black
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Richard M. Weinshilboum
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - John R. Hawse
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Thomas C. Spelsberg
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Matthew P. Goetz
- From the Departments of Oncology, Molecular Pharmacology and Experimental Therapeutics, Psychiatry, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| |
Collapse
|
20
|
Wang B, Yang LP, Zhang XZ, Huang SQ, Bartlam M, Zhou SF. New insights into the structural characteristics and functional relevance of the human cytochrome P450 2D6 enzyme. Drug Metab Rev 2010; 41:573-643. [PMID: 19645588 DOI: 10.1080/03602530903118729] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To date, the crystal structures of at least 12 human CYPs (1A2, 2A6, 2A13, 2C8, 2C9, 2D6, 2E1, 2R1, 3A4, 7A1, 8A1, and 46A1) have been determined. CYP2D6 accounts for only a small percentage of all hepatic CYPs (< 2%), but it metabolizes approximately 25% of clinically used drugs with significant polymorphisms. CYP2D6 also metabolizes procarcinogens and neurotoxins, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, 1,2,3,4-tetrahydroquinoline, and indolealkylamines. Moreover, the enzyme utilizes hydroxytryptamines and neurosteroids as endogenous substrates. Typical CYP2D6 substrates are usually lipophilic bases with an aromatic ring and a nitrogen atom, which can be protonated at physiological pH. Substrate binding is generally followed by oxidation (5-7 A) from the proposed nitrogen-Asp301 interaction. A number of homology models have been constructed to explore the structural features of CYP2D6, while antibody studies also provide useful structural information. Site-directed mutagenesis studies have demonstrated that Glu216, Asp301, Phe120, Phe481, and Phe483 play important roles in determining the binding of ligands to CYP2D6. The structure of human CYP2D6 has been recently determined and shows the characteristic CYP fold observed for other members of the CYP superfamily. The lengths and orientations of the individual secondary structural elements in the CYP2D6 structure are similar to those seen in other human CYP2 members, such as CYP2C9 and 2C8. The 2D6 structure has a well-defined active-site cavity located above the heme group with a volume of approximately 540 A(3), which is larger than equivalent cavities in CYP2A6 (260 A(3)), 1A2 (375 A(3)), and 2E1 (190 A(3)), but smaller than those in CYP3A4 (1385 A(3)) and 2C8 (1438 A(3)). Further studies are required to delineate the molecular mechanisms involved in CYP2D6 ligand interactions and their implications for drug development and clinical practice.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pediatrics, Guangdong Women and Children's Hospital, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
21
|
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
|
22
|
Yagihashi T, Mizuno M, Chino B, Sato Y, Sakuma K, Takebayashi T, Takao T, Kosaki K. Effects of the CYP2D6*10 alleles and co-medication with CYP2D6-dependent drugs on risperidone metabolism in patients with schizophrenia. Hum Psychopharmacol 2009; 24:301-8. [PMID: 19387994 DOI: 10.1002/hup.1025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Risperidone is converted to 9-hydroxyrisperidone by CYP2D6. Two parameters were used to examine the influences of CYP2D6 polymorphism and of co-medication on risperidone metabolism: the risperidone:9-hydroxyrisperidone concentration ratio (R:9-OHR ratio) and the sum of the risperidone and 9-hydroxyrisperidone concentrations divided by the dose (C:D ratio). We evaluated the effect of the CYP2D6*10 allele, which is a prevalent mutant allele among East Asians. METHODS Genotyping using the P450 microarray system was performed for 89 Japanese patients with schizophrenia receiving risperidone. The patients with CYP2D6*1/*1, *1/*2, or *2/*2 were classified as Group 1, those with one CYP2D6*10 allele (CYP2D6*1/*10 or *2/*10) were classified as Group 2, and those with two CYP2D6*10 alleles were classified as Group 3. The R:9-OHR and C:D ratios were analyzed using two-way ANOVAs with the CYP2D6 genotype and co-medication with CYP2D6-dependent drugs as independent variables. RESULTS Both the "genotype" and the "co-medication" factors had significant impacts on the R:9-OHR ratio (p = 0.011, p < 0.001). The "genotype" factor also had a significant impact on the C:D ratio (p = 0.032). However, the "co-medication" factor did not have a significant impact on the C:D ratio (p = 0.129). CONCLUSIONS The CYP2D6*10 polymorphism and the presence of co-medication exerted significant influences on the pharmacokinetics of risperidone.
Collapse
Affiliation(s)
- Tatsuhiko Yagihashi
- Department of Pediatrics, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Rae JM, Sikora MJ, Henry NL, Li L, Kim S, Oesterreich S, Skaar TC, Nguyen AT, Desta Z, Storniolo AM, Flockhart DA, Hayes DF, Stearns V. Cytochrome P450 2D6 activity predicts discontinuation of tamoxifen therapy in breast cancer patients. THE PHARMACOGENOMICS JOURNAL 2009; 9:258-64. [PMID: 19421167 DOI: 10.1038/tpj.2009.14] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The selective estrogen receptor modulator tamoxifen is routinely used for treatment and prevention of estrogen-receptor-positive breast cancer. Studies of tamoxifen adherence suggest that over half of patients discontinue treatment before the recommended 5 years. We hypothesized that polymorphisms in CYP2D6, the enzyme responsible for tamoxifen activation, predict for tamoxifen discontinuation. Tamoxifen-treated women (n=297) were genotyped for CYP2D6 variants and assigned a 'score' based on predicted allele activities from 0 (no activity) to 2 (high activity). Correlation between CYP2D6 score and discontinuation rates at 4 months was tested. We observed a strong nonlinear correlation between higher CYP2D6 score and increased rates of discontinuation (r(2)=0.935, P=0.018). These data suggest that presence of active CYP2D6 alleles may predict for higher likelihood of tamoxifen discontinuation. Therefore, patients who may be most likely to benefit from tamoxifen may paradoxically be most likely to discontinue treatment prematurely.
Collapse
Affiliation(s)
- J M Rae
- Breast Oncology Program, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109-0612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Johnson WW. Cytochrome P450 Inactivation by Pharmaceuticals and Phytochemicals: Therapeutic Relevance. Drug Metab Rev 2008; 40:101-47. [DOI: 10.1080/03602530701836704] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
25
|
Development of immobilized enzyme reactors based on human recombinant cytochrome P450 enzymes for phase I drug metabolism studies. J Chromatogr A 2008; 1206:2-10. [DOI: 10.1016/j.chroma.2008.05.080] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Revised: 05/26/2008] [Accepted: 05/27/2008] [Indexed: 11/21/2022]
|
26
|
Polasek TM, Miners JO. Time-dependent inhibition of human drug metabolizing cytochromes P450 by tricyclic antidepressants. Br J Clin Pharmacol 2008; 65:87-97. [PMID: 17662092 PMCID: PMC2291266 DOI: 10.1111/j.1365-2125.2007.02964.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Accepted: 04/17/2007] [Indexed: 11/29/2022] Open
Abstract
AIMS To investigate time-dependent inhibition (TDI) of human drug metabolizing CYP enzymes by tricyclic antidepressants (TCAs). METHODS CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A/CYP3A4 activities were investigated following co- and preincubation with TCAs using human liver microsomes (HLM) and human recombinant CYP proteins (expressed in Escherichia coli) as the enzyme sources. A two-step incubation method was employed to examine the in vitro mechanism-based inactivation (MBI) criteria. Potential metabolite-intermediate complex (MIC) formation was studied by spectral analysis. RESULTS TCAs generally exhibited significant TDI of recombinant CYP1A2, CYP2C19 and CYP2D6 (>10% positive inhibition differences between co- and preincubation conditions). TDI of recombinant CYP2C9 was minor (<10%), and was minor or absent in experiments utilizing recombinant CYP3A4 or HLM as the enzyme sources. Where observed, TDI of recombinant CYP occurred via alkylamine MIC formation, but evidence to support similar behaviour in HLM was limited. Indeed, only secondary amine TCAs reduced the apparent P450 content of HLM (3-6%) consistent with complexation. As a representative TCA, nortriptyline fulfilled the in vitro MBI criteria using recombinant CYP2C19 and CYP3A4 (K(I) and k(inact) values of 4 microm and 0.19 min(-1), and 70 microm and 0.06 min(-1)), but not with the human liver microsomal enzymes. CONCLUSIONS TCAs appear to have minimal potential for MBI of human liver microsomal CYP enzymes involved in drug metabolism. HLM and recombinant CYP (expressed in E. coli) are not equivalent enzyme sources for evaluating the TDI associated with some drugs.
Collapse
Affiliation(s)
- Thomas M Polasek
- Laboratory, Department of Clinical Pharmacology, Flinders University and Flinders Medical Centre, Adelaide, Australia.
| | | |
Collapse
|
27
|
Stadel R, Yang J, Nalwalk JW, Phillips JG, Hough LB. High-affinity binding of [3H]cimetidine to a heme-containing protein in rat brain. Drug Metab Dispos 2007; 36:614-21. [PMID: 18094038 DOI: 10.1124/dmd.107.017889] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
[(3)H]Cimetidine (3HCIM) specifically binds to an unidentified site in the rat brain. Because recently described ligands for this site have pharmacological activity, 3HCIM binding was characterized. 3HCIM binding was saturable, heat-labile, and distinct from the histamine H(2) receptor. To test the hypothesis that 3HCIM binds to a cytochrome P450 (P450), the effects of nonselective and isoform-selective P450 inhibitors were studied. The heme inhibitor KCN and the nonselective P450 inhibitor metyrapone both produced complete, concentration-dependent inhibition of 3HCIM binding (K(i) = 1.3 mM and 11.9 muM, respectively). Binding was largely unaffected by inhibitors of CYP1A2, 2B6, 2C8, 2C9, 2D6, 2E1, and 19A1 but was eliminated by inhibitors of CYP2C19 (tranylcypromine) and CYP3A4 (ketoconazole). Synthesis and testing of CC11 [4(5)-(benzylthiomethyl)-1H-imidazole] and CC12 [4(5)-((4-iodobenzyl)-thiomethyl)-1H-imidazole] confirmed both drugs to be high-affinity inhibitors of 3HCIM binding. On recombinant human P450s, CC12 was a potent inhibitor of CYP2B6 (IC(50) = 11.7 nM), CYP2C19 (51.4 nM), and CYP19A1 (140.7 nM) and had a range of activities (100-494 nM) on nine other isoforms. Although the 3HCIM binding site pharmacologically resembles some P450s, eight recombinant human P450s and three recombinant rat P450s did not exhibit 3HCIM binding. Inhibition by KCN and metyrapone suggests that 3HCIM binds to a heme-containing brain protein (possibly a P450). However, results with selective P450 inhibitors, recombinant P450 isoforms, and a P450 antibody did not identify a 3HCIM-binding P450 isoform. Finally, CC12 is a new, potent inhibitor of CYP2B6 and CYP2C19 that may be a valuable tool for P450 research.
Collapse
Affiliation(s)
- Rebecca Stadel
- Center for Neuropharmacology and Neuroscience, Albany Medical College MC-136, 47 New Scotland Ave, Albany, NY 12208, USA
| | | | | | | | | |
Collapse
|
28
|
Wu H, Yu W, Huang L, Wang J, Tang X, Yang W, Liu Y, Yu H, Zhu D. Effect of sodium ozagrel on the activity of rat CYP2D6. Eur J Pharmacol 2007; 573:55-9. [PMID: 17651725 DOI: 10.1016/j.ejphar.2007.06.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2006] [Revised: 06/13/2007] [Accepted: 06/18/2007] [Indexed: 11/23/2022]
Abstract
The aim of the study was to investigate the influence of sodium ozagrel on CYP2D6 (cytochromeP450 2D6) activity. The studies were performed with rat urine and liver microsomes and chemical inhibitors. The metabolism of dextromethorphan (dextrophan/dextromethorphan, dextrophan is a metabolite of dextromethorphan) and phenacetin (paracetamol/phenacetin, paracetamol is a metabolites of phenacetin) was used as probe to measure CYP2D6 and CYP1A2 (cytochromeP450 1A2) activity, respectively, determined by high-performance liquid chromatography (HPLC). The results showed that the metabolism of dextrophan/dextromethorphan in the sodium ozagrel-treated group (37 mg/kg) was higher than that of the control (P<0.05/6) in both in vivo and in vitro studies (r=0.9811). The rate of dextromethorphan metabolism was inhibited by sodium ozagrel and cimetidine in rat liver microsomes prepared from sodium ozagrel-treated rats and control rats group (sodium ozagrel IC(50)=26.5 microM, cimetidine IC(50)=86.3 microM in sodium ozagrel-treated group; sodium ozagrel IC(50)=13.9 microM, cimetidine IC(50)=24.8 microM in control group). The inhibitory effect of sodium ozagrel on CYP2D6 activity was noncompetitive with dextromethorphan with a K(i) of 324.94 microM. Kinetic parameters of the reactions were established by using Lineweaver-Burk with K(m)=0.67 mM and V(max)=2.13 pm/min/mg protein for the sodium ozagrel-treated group and K(m)=0.29 mM, and V(max)=0.91 pm/min/mg protein for the control group, respectively. The expression of CYP2D6 protein in the treated group was higher than that of the control group, as determined by Western blotting. The activity and expression of CYP1A2 did not show obvious differences in the control group and sodium ozagrel treated group. In conclusion, sodium ozagrel metabolism in rats is mediated primarily through CYP2D6, and sodium ozagrel can induce CYP2D6 activity.
Collapse
Affiliation(s)
- Hong Wu
- College of Pharmacy, Harbin Medical University, Harbin 150081, PR China; Mudanjiang Medical College, Mudanjiang 157011, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Li AP. In vitro evaluation of metabolic drug-drug interactions: a descriptive and critical commentary. CURRENT PROTOCOLS IN TOXICOLOGY 2007; Chapter 4:Unit 4.25. [PMID: 23045147 DOI: 10.1002/0471140856.tx0425s33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adverse drug-drug interactions represent a major challenge for the pharmaceutical industry. Recently, in vitro approaches for the evaluation of metabolism-related drug-drug interactions have been developed. These in vitro approaches are found to be useful in the assessment of clinical drug-drug interaction potential of new chemical entities and to aid the understanding of clinically significant drug-drug interactions observed with existing drugs. The general methods for the evaluation of drug-drug interactions using in vitro, human-based experimental systems are described and critically reviewed.
Collapse
Affiliation(s)
- Albert P Li
- In Vitro ADMET Laboratories, Columbia, Maryland, USA
| |
Collapse
|
30
|
Polasek TM, Miners JO. In vitroapproaches to investigate mechanism-based inactivation of CYP enzymes. Expert Opin Drug Metab Toxicol 2007; 3:321-9. [PMID: 17539741 DOI: 10.1517/17425255.3.3.321] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mechanism-based inactivation (MBI) of human drug-metabolising CYP enzymes is an important consideration in the preclinical ADME evaluation of new drug candidates. In this report, the in vitro approaches used to investigate MBI of CYP enzymes are described, with an emphasis on the characterisation required to assess potential drug-drug interactions. Recent disparities in MBI data between in vitro test systems are also reviewed, highlighting the limitations of Escherichia coli-expressed human recombinant CYP in the prediction of drug-drug interactions that arise via MBI.
Collapse
Affiliation(s)
- Thomas M Polasek
- Flinders University and Flinders Medical Centre, Department of Clinical Pharmacology, Bedford Park, 5042, Adelaide, Australia.
| | | |
Collapse
|
31
|
Takeda S, Kitajima Y, Ishii Y, Nishimura Y, Mackenzie PI, Oguri K, Yamada H. Inhibition of UDP-glucuronosyltransferase 2b7-catalyzed morphine glucuronidation by ketoconazole: dual mechanisms involving a novel noncompetitive mode. Drug Metab Dispos 2006; 34:1277-82. [PMID: 16679387 DOI: 10.1124/dmd.106.009738] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glucuronidation of morphine in humans is predominantly catalyzed by UDP-glucuronosyltransferase 2B7 (UGT2B7). Since our recent research suggested that cytochrome P450s (P450s) interact with UGT2B7 to affect its function [Takeda S et al. (2005) Mol Pharmacol 67:665-672], P450 inhibitors are expected to modulate UGT2B7-catalyzed activity. To address this issue, we investigated the effects of P450 inhibitors (cimetidine, sulfaphenazole, erythromycin, nifedipine, and ketoconazole) on the UGT2B7-catalyzed formation of morphine-3-glucuronide (M-3-G) and morphine-6-glucuronide (M-6-G). Among the inhibitors tested, ketoconazole was the most potent inhibitor of both M-3-G and M-6-G formation by human liver microsomes. The others were less effective except that nifedipine exhibited an inhibitory effect on M-6-G formation comparable to that by ketoconazole. Neither addition of NADPH nor solubilization of liver microsomes affected the ability of ketoconazole to inhibit morphine glucuronidation. In addition, ketoconazole had an ability to inhibit morphine UGT activity of recombinant UGT2B7 freed from P450. Kinetic analysis suggested that the ketoconazole-produced inhibition of morphine glucuronidation involves a mixed-type mechanism. Codeine potentiated inhibition of morphine glucuronidation by ketoconazole. In contrast, addition of another substrate, testosterone, showed no or a minor effect on ketoconazole-produced inhibition of morphine UGT. These results suggest that 1) metabolism of ketoconazole by P450 is not required for inhibition of UGT2B7-catalyzed morphine glucuronidation; and 2) this drug exerts its inhibitory effect on morphine UGT by novel mechanisms involving competitive and noncompetitive inhibition.
Collapse
Affiliation(s)
- Shuso Takeda
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|