1
|
Koroleva PI, Bulko TV, Agafonova LE, Shumyantseva VV. Catalytic and Electrocatalytic Mechanisms of Cytochromes P450 in the Development of Biosensors and Bioreactors. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1645-1657. [PMID: 38105030 DOI: 10.1134/s0006297923100176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 12/19/2023]
Abstract
Cytochromes P450 are a unique family of enzymes found in all Kingdoms of living organisms (animals, bacteria, plants, fungi, and archaea), whose main function is biotransformation of exogenous and endogenous compounds. The review discusses approaches to enhancing the efficiency of electrocatalysis by cytochromes P450 for their use in biotechnology and design of biosensors and describes main methods in the development of reconstituted and electrochemical catalytic systems based on the biochemical mechanism of cytochromes P450, as well as and modern trends for their practical application.
Collapse
Affiliation(s)
| | | | | | - Victoria V Shumyantseva
- Institute of Biomedical Chemistry, Moscow, 119121, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
2
|
Atypical kinetics of cytochrome P450 enzymes in pharmacology and toxicology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:131-176. [PMID: 35953154 DOI: 10.1016/bs.apha.2022.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atypical kinetics are observed in metabolic reactions catalyzed by cytochrome P450 enzymes (P450). Yet, this phenomenon is regarded as experimental artifacts in some instances despite increasing evidence challenging the assumptions of typical Michaelis-Menten kinetics. As P450 play a major role in the metabolism of a wide range of substrates including drugs and endogenous compounds, it becomes critical to consider the impact of atypical kinetics on the accuracy of estimated kinetic and inhibitory parameters which could affect extrapolation of pharmacological and toxicological implications. The first half of this book chapter will focus on atypical non-Michaelis-Menten kinetics (e.g. substrate inhibition, biphasic and sigmoidal kinetics) as well as proposed underlying mechanisms supported by recent insights in mechanistic enzymology. In particular, substrate inhibition kinetics in P450 as well as concurrent drug inhibition of P450 in the presence of substrate inhibition will be further discussed. Moreover, mounting evidence has revealed that despite the high degree of sequence homology between CYP3A isoforms (i.e. CYP3A4 and CYP3A5), they have the propensities to exhibit vastly different susceptibilities and potencies of mechanism-based inactivation (MBI) with a common drug inhibitor. These experimental observations pertaining to the presence of these atypical isoform- and probe substrate-specific complexities in CYP3A isoforms by several clinically-relevant drugs will therefore be expounded and elaborated upon in the second half of this book chapter.
Collapse
|
3
|
|
4
|
Kahma H, Aurinsalo L, Neuvonen M, Katajamäki J, Paludetto MN, Viinamäki J, Launiainen T, Filppula AM, Tornio A, Niemi M, Backman JT. An automated cocktail method for in vitro assessment of direct and time-dependent inhibition of nine major cytochrome P450 enzymes - application to establishing CYP2C8 inhibitor selectivity. Eur J Pharm Sci 2021; 162:105810. [PMID: 33753217 DOI: 10.1016/j.ejps.2021.105810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
We developed an in vitro high-throughput cocktail assay with nine major drug-metabolizing CYP enzymes, optimized for screening of time-dependent inhibition. The method was applied to determine the selectivity of the time-dependent CYP2C8 inhibitors gemfibrozil 1-O-β-glucuronide and clopidogrel acyl-β-D-glucuronide. In vitro incubations with CYP selective probe substrates and pooled human liver microsomes were conducted in 96-well plates with automated liquid handler techniques and metabolite concentrations were measured with quantitative UHPLC-MS/MS analysis. After determination of inter-substrate interactions and Km values for each reaction, probe substrates were divided into cocktails I (tacrine/CYP1A2, bupropion/CYP2B6, amodiaquine/CYP2C8, tolbutamide/CYP2C9 and midazolam/CYP3A4/5) and II (coumarin/CYP2A6, S-mephenytoin/CYP2C19, dextromethorphan/CYP2D6 and astemizole/CYP2J2). Time-dependent inhibitors (furafylline/CYP1A2, selegiline/CYP2A6, clopidogrel/CYP2B6, gemfibrozil 1-O-β-glucuronide/CYP2C8, tienilic acid/CYP2C9, ticlopidine/CYP2C19, paroxetine/CYP2D6 and ritonavir/CYP3A) and direct inhibitor (terfenadine/CYP2J2) showed similar inhibition with single substrate and cocktail methods. Established time-dependent inhibitors caused IC50 fold shifts ranging from 2.2 to 30 with the cocktail method. Under time-dependent inhibition conditions, gemfibrozil 1-O-β-glucuronide was a strong (>90% inhibition) and selective (<< 20% inhibition of other CYPs) inhibitor of CYP2C8 at concentrations ranging from 60 to 300 μM, while the selectivity of clopidogrel acyl-β-D-glucuronide was limited at concentrations above its IC80 for CYP2C8. The time-dependent IC50 values of these glucuronides for CYP2C8 were 8.1 and 38 µM, respectively. In conclusion, a reliable cocktail method including the nine most important drug-metabolizing CYP enzymes was developed, optimized and validated for detecting time-dependent inhibition. Moreover, gemfibrozil 1-O-β-glucuronide was established as a selective inhibitor of CYP2C8 for use as a diagnostic inhibitor in in vitro studies.
Collapse
Affiliation(s)
- Helinä Kahma
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Laura Aurinsalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jani Katajamäki
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marie-Noëlle Paludetto
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jenni Viinamäki
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Terhi Launiainen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Anne M Filppula
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
5
|
Tan BH, Ahemad N, Pan Y, Palanisamy UD, Othman I, Ong CE. In vitro inhibitory effects of glucosamine, chondroitin and diacerein on human hepatic CYP2D6. Drug Metab Pers Ther 2021; 36:259-270. [PMID: 34821124 DOI: 10.1515/dmpt-2020-0182] [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: 11/21/2020] [Accepted: 03/08/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Glucosamine, chondroitin and diacerein are natural compounds commonly used in treating osteoarthritis. Their concomitant intake may trigger drug-natural product interactions. Cytochrome P450 (CYP) has been implicated in such interactions. Cytochrome P450 2D6 (CYP2D6) is a major hepatic CYP involved in metabolism of 25% of the clinical drugs. This study aimed to investigate the inhibitory effect of these antiarthritic compounds on CYP2D6. METHODS CYP2D6 was heterologously expressed in Escherichia coli. CYP2D6-antiarthritic compound interactions were studied using in vitro enzyme kinetics assay and molecular docking. RESULTS The high-performance liquid chromatography (HPLC)-based dextromethorphan O-demethylase assay was established as CYP2D6 marker. All glucosamines and chondroitins weakly inhibited CYP2D6 (IC50 values >300 µM). Diacerein exhibited moderate inhibition with IC50 and K i values of 34.99 and 38.27 µM, respectively. Its major metabolite, rhein displayed stronger inhibition potencies (IC50=26.22 μM and K i =32.27 μM). Both compounds exhibited mixed-mode of inhibition. In silico molecular dockings further supported data from the in vitro study. From in vitro-in vivo extrapolation, rhein presented an area under the plasma concentration-time curve (AUC) ratio of 1.5, indicating low potential to cause in vivo inhibition. CONCLUSIONS Glucosamine, chondroitin and diacerein unlikely cause clinical interaction with the drug substrates of CYP2D6. Rhein, exhibits only low potential to cause in vivo inhibition.
Collapse
Affiliation(s)
- Boon Hooi Tan
- Division of Applied Biomedical Sciences and Biotechnology, International Medical University, Kuala Lumpur, Malaysia
| | - Nafees Ahemad
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Yan Pan
- Division of Biomedical Science, University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Uma Devi Palanisamy
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Chin Eng Ong
- School of Pharmacy, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Tan BH, Ahemad N, Pan Y, Palanisamy UD, Othman I, Ong CE. In vitro inhibitory effects of glucosamine, chondroitin and diacerein on human hepatic CYP2D6. Drug Metab Pers Ther 2021; 0:dmdi-2020-0182. [PMID: 33831979 DOI: 10.1515/dmdi-2020-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/08/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Glucosamine, chondroitin and diacerein are natural compounds commonly used in treating osteoarthritis. Their concomitant intake may trigger drug-natural product interactions. Cytochrome P450 (CYP) has been implicated in such interactions. Cytochrome P450 2D6 (CYP2D6) is a major hepatic CYP involved in metabolism of 25% of the clinical drugs. This study aimed to investigate the inhibitory effect of these antiarthritic compounds on CYP2D6. METHODS CYP2D6 was heterologously expressed in Escherichia coli. CYP2D6-antiarthritic compound interactions were studied using in vitro enzyme kinetics assay and molecular docking. RESULTS The high-performance liquid chromatography (HPLC)-based dextromethorphan O-demethylase assay was established as CYP2D6 marker. All glucosamines and chondroitins weakly inhibited CYP2D6 (IC50 values >300 µM). Diacerein exhibited moderate inhibition with IC50 and K i values of 34.99 and 38.27 µM, respectively. Its major metabolite, rhein displayed stronger inhibition potencies (IC50=26.22 μM and K i =32.27 μM). Both compounds exhibited mixed-mode of inhibition. In silico molecular dockings further supported data from the in vitro study. From in vitro-in vivo extrapolation, rhein presented an area under the plasma concentration-time curve (AUC) ratio of 1.5, indicating low potential to cause in vivo inhibition. CONCLUSIONS Glucosamine, chondroitin and diacerein unlikely cause clinical interaction with the drug substrates of CYP2D6. Rhein, exhibits only low potential to cause in vivo inhibition.
Collapse
Affiliation(s)
- Boon Hooi Tan
- Division of Applied Biomedical Sciences and Biotechnology, International Medical University, Kuala Lumpur, Malaysia
| | - Nafees Ahemad
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Yan Pan
- Division of Biomedical Science, University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Uma Devi Palanisamy
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Chin Eng Ong
- School of Pharmacy, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Schleiff MA, Flynn NR, Payakachat S, Schleiff BM, Pinson AO, Province DW, Swamidass SJ, Boysen G, Miller GP. Significance of Multiple Bioactivation Pathways for Meclofenamate as Revealed through Modeling and Reaction Kinetics. Drug Metab Dispos 2021; 49:133-141. [PMID: 33239334 PMCID: PMC7841419 DOI: 10.1124/dmd.120.000254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/05/2020] [Indexed: 12/20/2022] Open
Abstract
Meclofenamate is a nonsteroidal anti-inflammatory drug used in the treatment of mild-to-moderate pain yet poses a rare risk of hepatotoxicity through an unknown mechanism. Nonsteroidal anti-inflammatory drug (NSAID) bioactivation is a common molecular initiating event for hepatotoxicity. Thus, we hypothesized a similar mechanism for meclofenamate and leveraged computational and experimental approaches to identify and characterize its bioactivation. Analyses employing our XenoNet model indicated possible pathways to meclofenamate bioactivation into 19 reactive metabolites subsequently trapped into glutathione adducts. We describe the first reported bioactivation kinetics for meclofenamate and relative importance of those pathways using human liver microsomes. The findings validated only four of the many bioactivation pathways predicted by modeling. For experimental studies, dansyl glutathione was a critical trap for reactive quinone metabolites and provided a way to characterize adduct structures by mass spectrometry and quantitate yields during reactions. Of the four quinone adducts, we were able to characterize structures for three of them. Based on kinetics, the most efficient bioactivation pathway led to the monohydroxy para-quinone-imine followed by the dechloro-ortho-quinone-imine. Two very inefficient pathways led to the dihydroxy ortho-quinone and a likely multiply adducted quinone. When taken together, bioactivation pathways for meclofenamate accounted for approximately 13% of total metabolism. In sum, XenoNet facilitated prediction of reactive metabolite structures, whereas quantitative experimental studies provided a tractable approach to validate actual bioactivation pathways for meclofenamate. Our results provide a foundation for assessing reactive metabolite load more accurately for future comparative studies with other NSAIDs and drugs in general. SIGNIFICANCE STATEMENT: Meclofenamate bioactivation may initiate hepatotoxicity, yet common risk assessment approaches are often cumbersome and inefficient and yield qualitative insights that do not scale relative bioactivation risks. We developed and applied innovative computational modeling and quantitative kinetics to identify and validate meclofenamate bioactivation pathways and relevance as a function of time and concentration. This strategy yielded novel insights on meclofenamate bioactivation and provides a tractable approach to more accurately and efficiently assess other drug bioactivations and correlate risks to toxicological outcomes.
Collapse
Affiliation(s)
- Mary Alexandra Schleiff
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Noah R Flynn
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Sasin Payakachat
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Benjamin Mark Schleiff
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Anna O Pinson
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Dennis W Province
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - S Joshua Swamidass
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Gunnar Boysen
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| | - Grover P Miller
- Departments of Biochemistry and Molecular Biology (M.A.S, G.P.M.) and Environmental and Occupational Health (G.B.), University of Arkansas for Medical Sciences, Little Rock, Arizona (M.A.S.); Department of Pathology and Immunology, Washington University, St. Louis, Missouri (N.R.F., S.J.S.); Department of Chemistry, Hendrix College, Conway, Arizona (S.P.); and Independent Researcher (B.M.S.) and Department of Chemistry and Biochemistry (A.O.P., D.W.P.), Harding University, Searcy, Arkansas
| |
Collapse
|
8
|
Molecular probes for human cytochrome P450 enzymes: Recent progress and future perspectives. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213600] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Kahma H, Filppula AM, Launiainen T, Viinamäki J, Neuvonen M, Evangelista EA, Totah RA, Backman JT. Critical Differences between Enzyme Sources in Sensitivity to Detect Time-Dependent Inactivation of CYP2C8. Drug Metab Dispos 2019; 47:436-443. [PMID: 30709838 PMCID: PMC11022894 DOI: 10.1124/dmd.118.085498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Clopidogrel acyl-β-d-glucuronide is a mechanism-based inhibitor of cytochrome P450 2C8 in human liver microsomes (HLMs). However, time-dependent inactivation (TDI) of CYP2C8 could not be detected in an earlier study in human recombinant CYP2C8 (Supersomes). Here, we investigate whether different enzyme sources exhibit differences in detection of CYP2C8 TDI under identical experimental conditions. Inactivation of CYP2C8 by amiodarone (100 μM), clopidogrel acyl-β-d-glucuronide (100 μM), gemfibrozil 1-O-β-glucuronide (100 μM), and phenelzine (100 μM) was investigated in HLMs and three recombinant human CYP2C8 preparations (Supersomes, Bactosomes, and EasyCYP Bactosomes) using amodiaquine N-deethylation as the marker reaction. Furthermore, the inactivation kinetics of CYP2C8 by clopidogrel glucuronide (5-250 μM) was determined in Supersomes and Bactosomes. Amiodarone caused weak TDI in all enzyme preparations tested, while the extent of inactivation by clopidogrel glucuronide, gemfibrozil glucuronide, and phenelzine varied markedly between preparations, and even different Supersome lots. Both glucuronides caused strong inactivation of CYP2C8 in HLMs, Bactosomes and in one Supersome lot (>50% inhibition), but significant inactivation could not be reliably detected in other Supersome lots or EasyCYP Bactosomes. In Bactosomes, the concentration producing half of kinact (KI) and maximal inactivation rate (kinact) of clopidogrel glucuronide (14 μM and 0.054 minute-1) were similar to those determined previously in HLMs. Phenelzine caused strong inactivation of CYP2C8 in one Supersome lot (91% inhibition) but not in HLMs or other recombinant CYP2C8 preparations. In conclusion, different enzyme sources and different lots of the same recombinant enzyme preparation are not equally sensitive to detect inactivation of CYP2C8, suggesting that recombinant CYPs should be avoided when identifying mechanism-based inhibitors.
Collapse
Affiliation(s)
- Helinä Kahma
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Anne M Filppula
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Terhi Launiainen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Jenni Viinamäki
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Mikko Neuvonen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Eric A Evangelista
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Rheem A Totah
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| | - Janne T Backman
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, and Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland (H.K., A.M.F., T.L., J.V., M.N., J.T.B.) and Department of Medicinal Chemistry, University of Washington, Seattle, Washington (E.A.E., R.A.T.)
| |
Collapse
|
10
|
Qiu L, Li Q, Huang J, Wu Q, Tu K, Wu Y, Zhang X, Qian J, Zhang R, Li G, Sun M, Si L. In vitro effect of mPEG2k-PCLx micelles on rat liver cytochrome P450 enzymes. Int J Pharm 2018; 552:99-110. [PMID: 30253212 DOI: 10.1016/j.ijpharm.2018.09.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/03/2018] [Accepted: 09/20/2018] [Indexed: 02/06/2023]
|
11
|
Chen C, Liu J, Halpert JR, Wilderman PR. Use of Phenoxyaniline Analogues To Generate Biochemical Insights into the Interactio n of Polybrominated Diphenyl Ether with CYP2B Enzymes. Biochemistry 2018; 57:817-826. [PMID: 29215266 DOI: 10.1021/acs.biochem.7b01024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Human hepatic cytochromes P450 (CYP) are integral to xenobiotic metabolism. CYP2B6 is a major catalyst of biotransformation of environmental toxicants, including polybrominated diphenyl ethers (PBDEs). CYP2B substrates tend to contain halogen atoms, but the biochemical basis for this selectivity and for species specific determinants of metabolism has not been identified. Spectral binding titrations and inhibition studies were performed to investigate interactions of rat CYP2B1, rabbit CYP2B4, and CYP2B6 with a series of phenoxyaniline (POA) congeners that are analogues of PBDEs. For most congeners, there was a <3-fold difference between the spectral binding constants (KS) and IC50 values. In contrast, large discrepancies between these values were observed for POA and 3-chloro-4-phenoxyaniline. CYP2B1 was the enzyme most sensitive to POA congeners, so the Val-363 residue from that enzyme was introduced into CYP2B4 or CYP2B6. This substitution partially altered the protein-ligand interaction profiles to make them more similar to that of CYP2B1. Addition of cytochrome P450 oxidoreductase (POR) to titrations of CYP2B6 with POA or 2'4'5'TCPOA decreased the affinity of both ligands for the enzyme. Addition of cytochrome b5 to a recombinant enzyme system containing POR and CYP2B6 increased the POA IC50 value and decreased the 2'4'5'TCPOA IC50 value. Overall, the inconsistency between KS and IC50 values for POA versus 2'4'5'TCPOA is largely due to the effects of redox partner binding. These results provide insight into the biochemical basis of binding of diphenyl ethers to human CYP2B6 and changes in CYP2B6-mediated metabolism that are dependent on POA congener and redox partner identity.
Collapse
Affiliation(s)
- Chao Chen
- University of Connecticut School of Pharmacy , Storrs, Connecticut 06269, United States
| | - Jingbao Liu
- University of Connecticut School of Pharmacy , Storrs, Connecticut 06269, United States
| | - James R Halpert
- University of Connecticut School of Pharmacy , Storrs, Connecticut 06269, United States
| | - P Ross Wilderman
- University of Connecticut School of Pharmacy , Storrs, Connecticut 06269, United States
| |
Collapse
|
12
|
Wang S, Tang X, Yang T, Xu J, Zhang J, Liu X, Liu L. Predicted contributions of cytochrome P450s to drug metabolism in human liver microsomes using relative activity factor were dependent on probes. Xenobiotica 2018; 49:161-168. [PMID: 29375004 DOI: 10.1080/00498254.2018.1433902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Contributions of cytochrome P450 (CYP450) isoforms to drug metabolism are often predicted using relative activity factor (RAF) method, assuming RAF values were independent of probe. We aimed to report probe-dependent characteristic of RAF values using CYP3A4 or CYP2C9 probes. Metabolism of four CYP3A4 probes (testosterone, midazolam, verapamil and atorvastatin) and three CYP2C9 probes (tolbutamide, diclofenac and S-warfarin) in human liver microsomes (HLM) and cDNA-expressed recombinant CYP450 (Rec-CYP450) systems were characterized and RAFCL value was estimated as ratio of probe intrinsic clearance in HLM to that in Rec-CYP450. CYP450i contributions to metabolic reaction of a probe were predicted using other probes and compared with data from specific inhibitions. Contributions of CYP3A4 and CYP2C9 to metabolism of deoxypodophyllotoxin and nateglinide were also predicted. RAF values were dependent on probes, leading to probe-dependently predicted contributions. Predicted contributions of CYP3A4 to formations of 6β-hydroxytestosterone, 1'-hydroxymidazolam, norverapamil, ortho-hydroxyatorvastatin and para-hydroxyatorvastatin using other probes were 47.46-219.46%, 21.62-98.87%, 186.49-462.44%, 21.87-101.15% and 53.62-247.97%, respectively. Predicted contributions of CYP3A4 and CYP2C9 to nateglinide metabolism were 8.18-37.84% and 36.08-94.04%, separately. In conclusion, CYP450i contribution to drug metabolism in HLM estimated using RAF approach were probe-dependent. Therefore, contribution of each isoform must be confirmed by multiple probes.
Collapse
Affiliation(s)
- Shuting Wang
- a Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Xiange Tang
- a Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Tingting Yang
- a Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Jiong Xu
- a Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Jiaxin Zhang
- a Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Xiaodong Liu
- a Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Li Liu
- a Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
13
|
Effects of dronedarone, amiodarone and their active metabolites on sequential metabolism of arachidonic acid to epoxyeicosatrienoic and dihydroxyeicosatrienoic acids. Biochem Pharmacol 2017; 146:188-198. [DOI: 10.1016/j.bcp.2017.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/22/2017] [Indexed: 12/23/2022]
|
14
|
Effect of ketoconazole on the transport and metabolism of drugs in the human liver cell model. Russ Chem Bull 2017. [DOI: 10.1007/s11172-017-1713-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
15
|
Venkatachalam A, Parashar A, Manoj KM. Functioning of drug-metabolizing microsomal cytochrome P450s: In silico probing of proteins suggests that the distal heme 'active site' pocket plays a relatively 'passive role' in some enzyme-substrate interactions. In Silico Pharmacol 2016; 4:2. [PMID: 26894412 PMCID: PMC4760962 DOI: 10.1186/s40203-016-0016-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/05/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE The currently held mechanistic understanding of microsomal cytochrome P450s (CYPs) seeks that diverse drug molecules bind within the deep-seated distal heme pocket and subsequently react at the heme centre. To explain a bevy of experimental observations and meta-analyses, we indulge a hypothesis that involves a "diffusible radical mediated" mechanism. This new hypothesis posits that many substrates could also bind at alternate loci on/within the enzyme and be reacted without the pertinent moiety accessing a bonding proximity to the purported catalytic Fe-O enzyme intermediate. METHODS Through blind and heme-distal pocket centered dockings of various substrates and non-substrates (drug molecules of diverse sizes, classes, topographies etc.) of microsomal CYPs, we explored the possibility of access of substrates via the distal channels, its binding energies, docking orientations, distance of reactive moieties (or molecule per se) to/from the heme centre, etc. We investigated specific cases like- (a) large drug molecules as substrates, (b) classical marker drug substrates, (c) class of drugs as substrates (Sartans, Statins etc.), (d) substrate preferences between related and unrelated CYPs, (e) man-made site-directed mutants' and naturally occurring mutants' reactivity and metabolic disposition, (f) drug-drug interactions, (g) overall affinities of drug substrate versus oxidized product, (h) meta-analysis of in silico versus experimental binding constants and reaction/residence times etc. RESULTS It was found that heme-centered dockings of the substrate/modulator drug molecules with the available CYP crystal structures gave poor docking geometries and distances from Fe-heme centre. In conjunction with several other arguments, the findings discount the relevance of erstwhile hypothesis in many CYP systems. Consequently, the newly proposed hypothesis is deemed a viable alternate, as it satisfies Occam's razor. CONCLUSIONS The new proposal affords expanded scope for explaining the mechanism, kinetics and overall phenomenology of CYP mediated drug metabolism. It is now understood that the heme-iron and the hydrophobic distal pocket of CYPs serve primarily to stabilize the reactive intermediate (diffusible radical) and the surface or crypts of the apoprotein bind to the xenobiotic substrate (and in some cases, the heme distal pocket could also serve the latter function). Thus, CYPs enhance reaction rates and selectivity/specificity via a hitherto unrecognized modality.
Collapse
Affiliation(s)
- Avanthika Venkatachalam
- Formerly at PSG Institute of Advanced Studies, Avinashi Road, Peelamedu, Coimbatore, Tamil Nadu, 641004, India.
| | - Abhinav Parashar
- Formerly at Hemoproteins Lab, School of Bio Sciences and Technology, VIT University, Vellore, Tamil Nadu, India, 632014.
| | - Kelath Murali Manoj
- Formerly at PSG Institute of Advanced Studies, Avinashi Road, Peelamedu, Coimbatore, Tamil Nadu, 641004, India.
- Formerly at Hemoproteins Lab, School of Bio Sciences and Technology, VIT University, Vellore, Tamil Nadu, India, 632014.
- Satyamjayatu: The Science & Ethics Foundation, Kulappully, Shoranur-2 (PO), Kerala, 679122, India.
| |
Collapse
|
16
|
Parmentier Y, Pothier C, Delmas A, Caradec F, Trancart MM, Guillet F, Bouaita B, Chesne C, Brian Houston J, Walther B. Direct and quantitative evaluation of the human CYP3A4 contribution (fm) to drug clearance using the in vitro SILENSOMES model. Xenobiotica 2016; 47:562-575. [DOI: 10.1080/00498254.2016.1208854] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yannick Parmentier
- Biopharmaceutical Research Department, Technologie Servier, Orléans Cedex, France,
| | - Corinne Pothier
- Biopharmaceutical Research Department, Technologie Servier, Orléans Cedex, France,
| | - Audrey Delmas
- Biopharmaceutical Research Department, Technologie Servier, Orléans Cedex, France,
| | - Fabrice Caradec
- Biopharmaceutical Research Department, Technologie Servier, Orléans Cedex, France,
| | | | | | | | | | | | - Bernard Walther
- Biopharmaceutical Research Department, Technologie Servier, Orléans Cedex, France,
| |
Collapse
|
17
|
den Braver-Sewradj SP, den Braver MW, Vermeulen NP, Commandeur JN, Richert L, Vos JC. Inter-donor variability of phase I/phase II metabolism of three reference drugs in cryopreserved primary human hepatocytes in suspension and monolayer. Toxicol In Vitro 2016; 33:71-9. [DOI: 10.1016/j.tiv.2016.02.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/18/2016] [Accepted: 02/21/2016] [Indexed: 12/20/2022]
|
18
|
Xie Q, Chen Y, Liu F, Zhong Z, Zhao K, Ling Z, Wang F, Tang X, Wang Z, Liu L, Liu X. Interspecies differences in metabolism of deoxypodophyllotoxin in hepatic microsomes from human, monkey, rat, mouse and dog. Drug Metab Pharmacokinet 2016; 31:314-22. [PMID: 27329261 DOI: 10.1016/j.dmpk.2016.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 01/11/2023]
Abstract
Deoxypodophyllotoxin (DPT) is a natural lignan product which has drawn much attention due to its pharmacological properties including antitumor effect. The purpose of this study was to investigate interspecies differences in metabolism of DPT in hepatic microsomes from human (HLM), cynomolgus monkey (CyLM), rat (RLM), mouse (MLM) and dog (DLM). Incubation of DPT with hepatic microsomes from five species in the presence of NADPH resulted in formation of seven metabolites, five of which were compared with the synthetic standards. M2 was the most abundant metabolite in microsomes from all species. Rank order of intrinsic clearance for M2 formation was RLM > CyLM > MLM > HLM > DLM. In HLM, sulfaphenazole showed the strongest inhibition effect on M2 formation, but neither ticlopidine nor ketoconazole inhibited M2 formation in HLM. Results from cDNA-expressed human CYP450s experiments showed that clearance of M2 formation was much higher in CYP2C9 and CYP2C19 than that in CYP3A4. Contributions of the three CYP450 isoforms to M2 formation in HLM were estimated using relative activity factor (RAF) method or correction by amount of CYP450 isoforms in HLM. M2 formation in HLM was mainly attributed to CYP2C9, followed by CYP2C19. Involvement of CYP3A4 was minor.
Collapse
Affiliation(s)
- Qiushi Xie
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Chen
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Fei Liu
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Zeyu Zhong
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Kaijing Zhao
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Zhaoli Ling
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Wang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Xiange Tang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongjian Wang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
19
|
Schejbal J, Řemínek R, Zeman L, Mádr A, Glatz Z. On-line coupling of immobilized cytochrome P450 microreactor and capillary electrophoresis: A promising tool for drug development. J Chromatogr A 2016; 1437:234-240. [DOI: 10.1016/j.chroma.2016.01.081] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/13/2016] [Accepted: 01/29/2016] [Indexed: 10/22/2022]
|
20
|
Lu Z, Ma G, Veinot JGC, Wong CS. Disruption of biomolecule function by nanoparticles: how do gold nanoparticles affect Phase I biotransformation of persistent organic pollutants? CHEMOSPHERE 2013; 93:123-32. [PMID: 23763865 DOI: 10.1016/j.chemosphere.2013.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 04/23/2013] [Accepted: 05/02/2013] [Indexed: 05/22/2023]
Abstract
The potential influence of nanoparticles on cytochrome P-450 (CYP) isozyme mediated Phase I biotransformation of persistent organic pollutants (POPs) in vitro was investigated using citrate-capped gold nanoparticles (AuNPs) and 2,2',3,5',6-pentachlorobiphenyl (PCB 95) as the probe nanoparticle and compound, respectively. AuNPs affected the biotransformation activity of rat CYP2B1 and changed the atropisomeric composition of PCB 95, depending on the incubation time and the AuNP concentration. Electrostatic repulsion between citrate-coated AuNPs and rat CYP2B1 may influence the active conformation of the isozyme and consequently affect its activity and stereoselectivity. In addition, the effects of AuNPs on rat CYP2B1 activity also appeared to be through interference with the CYP catalytic cycle's electron transfer chain. Incubations with AuNPs had a decline in buffer conductance and an absorbance band red shift of AuNPs, from electrostatic interactions of K(+) with negatively-charged AuNP aggregates. These ionic strength changes affected the formation rate of nicotinamide adenine dinucleotide phosphate, which provides electrons for the oxidative reaction cycle, and the biotransformation activity and stereoselectivity of CYP. This study suggests that charged nanoparticles may be able to alter the functions of biomolecules directly, by electrostatic interaction, or indirectly, by changes to the surrounding ionic strength. These factors should be taken into account for further understanding and prediction of the environmental behavior and fate of POPs and nanoparticles.
Collapse
Affiliation(s)
- Zhe Lu
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada R3T 2N2
| | | | | | | |
Collapse
|
21
|
Ong CE, Pan Y, Mak JW, Ismail R. In vitro approaches to investigate cytochrome P450 activities: update on current status and their applicability. Expert Opin Drug Metab Toxicol 2013; 9:1097-113. [PMID: 23682848 DOI: 10.1517/17425255.2013.800482] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Cytochromes P450 (CYPs) play a central role in the Phase I metabolism of drugs and other xenobiotics. It is estimated that CYPs can metabolize up to two-thirds of drugs present in humans. Over the past two decades, there have been numerous advances in in vitro methodologies to characterize drug metabolism and interaction involving CYPs. AREAS COVERED This review focuses on the use of in vitro methodologies to examine CYPs' role in drug metabolism and interaction. There is an emphasis on their current development, applicability, advantages and limitations as well as the use of in silico approaches in complementing and supporting in vitro data. The article also highlights the challenges in extrapolating in vitro data to in vivo situations. EXPERT OPINION Advances in in vitro methodologies have been made such that data can be used for in vivo prediction with comfortable degree of confidence. Improved assay designs and analytical techniques have permitted development of miniaturized assay format and automated system with improved sensitivity and throughput capacity. High-quality experimental designs and scientifically rigorous assessment/validation protocols remain crucial in developing reliable and robust in vitro models. With continued progress made in the field, in vitro methodologies will continually be employed in evaluating CYP activities in pharmaceutical industries and laboratories.
Collapse
Affiliation(s)
- Chin Eng Ong
- Monash University Sunway Campus, Jeffrey Cheah School of Medicine and Health Sciences, Jalan Lagoon Selatan, 46150 Bandar Sunway, Selangor, Malaysia.
| | | | | | | |
Collapse
|
22
|
Yan Z, Caldwell GW. In vitro identification of cytochrome P450 enzymes responsible for drug metabolism. Methods Mol Biol 2013; 1015:251-261. [PMID: 23824861 DOI: 10.1007/978-1-62703-435-7_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Metabolism catalyzed by the cytochrome P450 enzymes (CYPs) represents the most important pathway for drug metabolism and elimination in humans. Identification of the CYPs responsible for metabolism of existing and novel drugs is critical for the prediction of adverse reactions caused by drug-drug interactions or individual genetic polymorphism. An integrated approach is described for CYP-mediated metabolic reaction phenotyping using both recombinant enzymes and human liver microsomes in combination of selective inhibitors or inhibitory antibodies. The in vitro method described includes screening of recombinant CYPs for metabolic activity, chemical inhibition or antibody neutralization, and correlation analysis with isoform-selective marker activities. The primary focus is on identification of the most common enzymes including CYP1A2, 2C9, 2C19, 2D6, and 3A4, although the same strategy could potentially be used for identification of other isoforms.
Collapse
Affiliation(s)
- Zhengyin Yan
- CREATe, Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | | |
Collapse
|
23
|
González-Pérez V, Connolly EA, Bridges AS, Wienkers LC, Paine MF. Impact of organic solvents on cytochrome P450 probe reactions: filling the gap with (S)-Warfarin and midazolam hydroxylation. Drug Metab Dispos 2012; 40:2136-42. [PMID: 22896727 PMCID: PMC3477202 DOI: 10.1124/dmd.112.047134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/14/2012] [Indexed: 12/18/2022] Open
Abstract
(S)-Warfarin 7-hydroxylation and midazolam 1'-hydroxylation are among the preferred probe substrate reactions for CYP2C9 and CYP3A4/5, respectively. The impact of solvents on enzyme activity, kinetic parameters, and predicted in vivo hepatic clearance (Cl(H)) associated with each reaction has not been evaluated. The effects of increasing concentrations [0.1-2% (v/v)] of six organic solvents (acetonitrile, methanol, ethanol, dimethyl sulfoxide, acetone, isopropanol) were first tested on each reaction using human liver microsomes (HLMs), human intestinal microsomes (midazolam 1'-hydroxylation only), and recombinant enzymes. Across enzyme sources, relative to water, acetonitrile and methanol had the least inhibitory effect on (S)-warfarin 7-hydroxylation (0-58 and 9-96%, respectively); acetonitrile, methanol, and ethanol had the least inhibitory effect on midazolam 1'-hydroxylation (0-29, 0-22, and 0-20%, respectively). Using HLMs, both acetonitrile and methanol (0.1-2%) decreased the V(max) (32-60 and 24-65%, respectively) whereas methanol (2%) increased the K(m) (100%) of (S)-warfarin-hydroxylation. (S)-Warfarin Cl(H) was underpredicted by 21-65% (acetonitrile) and 13-84% (methanol). Acetonitrile, methanol, and ethanol had minimal to modest impact on both the kinetics of midazolam 1'-hydroxylation (10-24%) and predicted midazolam Cl(H) (2-20%). In conclusion, either acetonitrile or methanol at ≤0.1% is recommended as the primary organic solvent for the (S)-warfarin 7-hydroxylation reaction; acetonitrile is preferred if higher solvent concentrations are required. Acetonitrile, methanol, and ethanol at ≤2% are recommended as primary organic solvents for the midazolam 1'-hydroxylation reaction. This information should facilitate optimization of experimental conditions and improve the interpretation and accuracy of in vitro-in vivo predictions involving these two preferred cytochrome P450 probe substrate reactions.
Collapse
Affiliation(s)
- Vanessa González-Pérez
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
24
|
Wollenberg LA, Jett JE, Wu Y, Flora DR, Wu N, Tracy TS, Gannett PM. Selective filling of nanowells in nanowell arrays fabricated using polystyrene nanosphere lithography with cytochrome P450 enzymes. NANOTECHNOLOGY 2012; 23:385101. [PMID: 22947619 PMCID: PMC3465080 DOI: 10.1088/0957-4484/23/38/385101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
This work describes an original and simple technique for protein immobilization into nanowells, fabricated using nanopatterned array fabrication methods, while ensuring the protein retains normal biological activity. Nanosphere lithography was used to fabricate a nanowell array with nanowells 100 nm in diameter with a periodicity of 500 nm. The base of the nanowells was gold and the surrounding material was silicon dioxide. The different surface chemistries of these materials were used to attach two different self-assembled monolayers (SAM) with different affinities for the protein used here, cytochrome P450 (P450). The nanowell SAM, a methyl terminated thiol, had high affinity for the P450. The surrounding SAM, a polyethylene glycol silane, displayed very little affinity toward the P450 isozyme CYP2C9, as demonstrated by x-ray photoelectron spectroscopy and surface plasmon resonance. The regularity of the nanopatterned array was examined by scanning electron microscopy and atomic force microscopy. P450-mediated metabolism experiments of known substrates demonstrated that the nanowell bound P450 enzyme exceeded its normal activity, as compared to P450 solutions, when bound to the methyl terminated self-assembled monolayer. The nanopatterned array chips bearing P450 display long term stability and give reproducible results making them potentially useful for high-throughput screening assays or as nanoelectrode arrays.
Collapse
Affiliation(s)
- Lance A. Wollenberg
- West Virginia University, Basic Pharmaceutical Sciences, P.O. Box 9530, Morgantown, WV 26506
| | - John E. Jett
- West Virginia University, Basic Pharmaceutical Sciences, P.O. Box 9530, Morgantown, WV 26506
| | - Yueting Wu
- West Virginia University, Department of Chemical Engineering, P.O. Box 6102, Morgantown, WV 26506
| | - Darcy R. Flora
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455
| | - Nianqiang Wu
- West Virginia University, Department of Chemical Engineering, P.O. Box 6102, Morgantown, WV 26506
| | - Timothy S. Tracy
- University of Kentucky, College of Pharmacy, 789 S. Limestone, Lexington, KY 40536
| | - Peter M. Gannett
- West Virginia University, Basic Pharmaceutical Sciences, P.O. Box 9530, Morgantown, WV 26506
| |
Collapse
|
25
|
Darnell M, Ulvestad M, Ellis E, Weidolf L, Andersson TB. In Vitro Evaluation of Major In Vivo Drug Metabolic Pathways Using Primary Human Hepatocytes and HepaRG Cells in Suspension and a Dynamic Three-Dimensional Bioreactor System. J Pharmacol Exp Ther 2012; 343:134-44. [DOI: 10.1124/jpet.112.195834] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
26
|
Quintieri L, Palatini P, Moro S, Floreani M. Inhibition of cytochrome P450 2C8-mediated drug metabolism by the flavonoid diosmetin. Drug Metab Pharmacokinet 2012; 26:559-68. [PMID: 21791871 DOI: 10.2133/dmpk.dmpk-11-rg-048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The aim of this study was to assess the effects of diosmetin and hesperetin, two flavonoids present in various medicinal products, on CYP2C8 activity of human liver microsomes using paclitaxel oxidation to 6α-hydroxy-paclitaxel as a probe reaction. Diosmetin and hesperetin inhibited 6α-hydroxy-paclitaxel production in a concentration-dependent manner, diosmetin being about 16-fold more potent than hesperetin (mean IC(50) values 4.25 ± 0.02 and 68.5 ± 3.3 µM for diosmetin and hesperetin, respectively). Due to the low inhibitory potency of hesperetin, we characterized the mechanism of diosmetin-induced inhibition only. This flavonoid proved to be a reversible, dead-end, full inhibitor of CYP2C8, its mean inhibition constant (K(i)) being 3.13 ± 0.11 µM. Kinetic analysis showed that diosmetin caused mixed-type inhibition, since it significantly decreased the V(max) (maximum velocity) and increased the K(m) value (substrate concentration yielding 50% of V(max)) of the reaction. The results of kinetic analyses were consistent with those of molecular docking simulation, which showed that the putative binding site of diosmetin coincided with the CYP2C8 substrate binding site. The demonstration that diosmetin inhibits CYP2C8 at concentrations similar to those observed after in vivo administration (in the low micromolar range) is of potential clinical relevance, since it may cause pharmacokinetic interactions with co-administered drugs metabolized by this CYP.
Collapse
Affiliation(s)
- Luigi Quintieri
- Department of Pharmacology and Anaesthesiology, University of Padova, Italy
| | | | | | | |
Collapse
|
27
|
Xu C, Ogburn ET, Guo Y, Desta Z. Effects of the CYP2B6*6 allele on catalytic properties and inhibition of CYP2B6 in vitro: implication for the mechanism of reduced efavirenz metabolism and other CYP2B6 substrates in vivo. Drug Metab Dispos 2012; 40:717-25. [PMID: 22232427 PMCID: PMC3310426 DOI: 10.1124/dmd.111.042416] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 01/09/2012] [Indexed: 01/11/2023] Open
Abstract
The mechanism by which CYP2B6*6 allele alters drug metabolism in vitro and in vivo is not fully understood. To test the hypothesis that altered substrate binding and/or catalytic properties contribute to its functional consequences, efavirenz 8-hydroxylation and bupropion 4-hydroxylation were determined in CYP2B6.1 and CYP2B6.6 proteins expressed without and with cytochrome b5 (Cyt b5) and in human liver microsomes (HLMs) obtained from liver tissues genotyped for the CYP2B6*6 allele. The susceptibility of the variant protein to inhibition was also tested in HLMs. Significantly higher V(max) and K(m) values for 8-hydroxyefavirenz formation and ∼2-fold lower intrinsic clearance (Cl(int)) were noted in expressed CYP2B6.6 protein (-b5) compared with that of CYP2B6.1 protein (-b5); this effect was abolished by Cyt b5. The V(max) and Cl(int) values for 4-hydroxybupropion formation were significantly higher in CYP2B6.6 than in CYP2B6.1 protein, with no difference in K(m), whereas coexpression with Cyt b5 reversed the genetic effect on these kinetic parameters. In HLMs, CYP2B6*6/*6 genotype was associated with markedly lower V(max) (and moderate increase in K(m)) and thus lower Cl(int) values for efavirenz and bupropion metabolism, but no difference in catalytic properties was noted between CYP2B6*1/*1 and CYP2B6*1/*6 genotypes. Inhibition of efavirenz 8-hydroxylation by voriconazole was significantly greater in HLMs with the CYP2B6*6 allele (K(i) = 1.6 ± 0.8 μM) than HLMs with CYP2B6*1/*1 genotype (K(i) = 3.0 ± 1.1 μM). In conclusion, our data suggest the CYP2B6*6 allele influences metabolic activity by altering substrate binding and catalytic activity in a substrate- and Cyt b5-dependent manner. It may also confer susceptibility to inhibition.
Collapse
Affiliation(s)
- Cong Xu
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
28
|
Vestergren AS, Zlabek V, Pickova J, Zamaratskaia G. Tolbutamide hydroxylation by hepatic microsomes from Atlantic salmon (Salmo salar L.). Mol Biol Rep 2012; 39:6867-73. [DOI: 10.1007/s11033-012-1512-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/24/2012] [Indexed: 11/30/2022]
|
29
|
Raccor BS, Claessens AJ, Dinh JC, Park JR, Hawkins DS, Thomas SS, Makar KW, McCune JS, Totah RA. Potential contribution of cytochrome P450 2B6 to hepatic 4-hydroxycyclophosphamide formation in vitro and in vivo. Drug Metab Dispos 2012; 40:54-63. [PMID: 21976622 PMCID: PMC3250049 DOI: 10.1124/dmd.111.039347] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 10/03/2011] [Indexed: 01/11/2023] Open
Abstract
Results from retrospective studies on the relationship between cytochrome P450 (P450) 2B6 (CYP2B6) genotype and cyclophosphamide (CY) efficacy and toxicity in adult cancer patients have been conflicting. We evaluated this relationship in children, who have faster CY clearance and receive different CY-based regimens than adults. These factors may influence the P450s metabolizing CY to 4-hydroxycyclophosphamide (4HCY), the principal precursor to CY's cytotoxic metabolite. Therefore, we sought to characterize the in vitro and in vivo roles of hepatic CYP2B6 and its main allelic variants in 4HCY formation. CYP2B6 is the major isozyme responsible for 4HCY formation in recombinant P450 Supersomes. In human liver microsomes (HLM), 4HCY formation correlated with known phenotypic markers of CYP2B6 activity, specifically formation of (S)-2-ethyl-1,5-dimethyl-3,3-diphenyl pyrrolidine and hydroxybupropion. However, in HLM, CYP3A4/5 also contributes to 4HCY formation at the CY concentrations similar to plasma concentrations achieved in children (0.1 mM). 4HCY formation was not associated with CYP2B6 genotype at low (0.1 mM) or high (1 mM) CY concentrations potentially because CYP3A4/5 and other isozymes also form 4HCY. To remove this confounder, 4HCY formation was evaluated in recombinant CYP2B6 enzymes, which demonstrated that 4HCY formation was lower for CYP2B6.4 and CYP2B6.5 compared with CYP2B6.1. In vivo, CYP2B6 genotype was not directly related to CY clearance or ratio of 4HCY/CY areas under the curve in 51 children receiving CY-based regimens. Concomitant chemotherapy agents did not influence 4HCY formation in vitro. We conclude that CYP2B6 genotype is not consistently related to 4HCY formation in vitro or in vivo.
Collapse
Affiliation(s)
- Brianne S Raccor
- Department of Pharmacy, University of Washington, Seattle, WA 98195-7610, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Traylor MJ, Ryan JD, Arnon ES, Dordick JS, Clark DS. Rapid and quantitative measurement of metabolic stability without chromatography or mass spectrometry. J Am Chem Soc 2011; 133:14476-9. [PMID: 21766815 DOI: 10.1021/ja203172c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Metabolic stability measurements are a critical component of preclinical drug development. Available measurement strategies rely on chromatography and mass spectrometry, which are expensive and labor intensive. We have developed a general method to determine the metabolic stability of virtually any compound by quantifying cofactors in the mechanism of cytochrome P450 enzymes using fluorescence intensity measurements. While many previous studies have shown that simple measurements of cofactor depletion do not correlate with substrate conversion (i.e., metabolic stability) in P450 systems, the present work employs a reaction engineering approach to simplify the overall rate equation, thus allowing the accurate and quantitative determination of substrate depletion from simultaneous measurements of NADPH and oxygen depletion. This method combines the accuracy and generality of chromatography with the ease, throughput, and real-time capabilities of fluorescence.
Collapse
Affiliation(s)
- Matthew J Traylor
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | | | | | | | | |
Collapse
|
31
|
Crewe HK, Barter ZE, Yeo KR, Rostami-Hodjegan A. Are there differences in the catalytic activity per unit enzyme of recombinantly expressed and human liver microsomal cytochrome P450 2C9? A systematic investigation into inter-system extrapolation factors. Biopharm Drug Dispos 2011; 32:303-18. [PMID: 21725985 DOI: 10.1002/bdd.760] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 05/02/2011] [Accepted: 05/05/2011] [Indexed: 11/07/2022]
Abstract
The 'relative activity factor' (RAF) compares the activity per unit of microsomal protein in recombinantly expressed cytochrome P450 enzymes (rhCYP) and human liver without separating the potential sources of variation (i.e. abundance of enzyme per mg of protein or variation of activity per unit enzyme). The dimensionless 'inter-system extrapolation factor' (ISEF) dissects differences in activity from those in CYP abundance. Detailed protocols for the determination of this scalar, which is used in population in vitro-in vivo extrapolation (IVIVE), are currently lacking. The present study determined an ISEF for CYP2C9 and, for the first time, systematically evaluated the effects of probe substrate, cytochrome b5 and methods for assessing the intrinsic clearance (CL(int) ). Values of ISEF for S-warfarin, tolbutamide and diclofenac were 0.75 ± 0.18, 0.57 ± 0.07 and 0.37 ± 0.07, respectively, using CL(int) values derived from the kinetic values V(max) and K(m) of metabolite formation in rhCYP2C9 + reductase + b5 BD Supersomes™. The ISEF values obtained using rhCYP2C9 + reductase BD Supersomes™ were more variable, with values of 7.16 ± 1.25, 0.89 ± 0.52 and 0.50 ± 0.05 for S-warfarin, tolbutamide and diclofenac, respectively. Although the ISEF values obtained from rhCYP2C9 + reductase + b5 for the three probe substrates were statistically different (p < 0.001), the use of the mean value of 0.54 resulted in predicted oral clearance values for all three substrates within 1.4 fold of the observed literature values. For consistency in the relative activity across substrates, use of a b5 expressing recombinant system, with the intrinsic clearance calculated from full kinetic data is recommended for generation of the CYP2C9 ISEF. Furthermore, as ISEFs have been found to be sensitive to differences in accessory proteins, rhCYP system specific ISEFs are recommended.
Collapse
Affiliation(s)
- H K Crewe
- Academic Unit of Clinical Pharmacology, University of Sheffield, Royal Hallamshire Hospital, Sheffield, UK
| | | | | | | |
Collapse
|
32
|
Rossato G, Ernst B, Smiesko M, Spreafico M, Vedani A. Probing small-molecule binding to cytochrome P450 2D6 and 2C9: An in silico protocol for generating toxicity alerts. ChemMedChem 2011; 5:2088-101. [PMID: 21038340 DOI: 10.1002/cmdc.201000358] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Drug metabolism, toxicity, and their interaction profiles are major issues in the drug-discovery and lead-optimization processes. The cytochromes P450 (CYPs) 2D6 and 2C9 are enzymes involved in the oxidative metabolism of a majority of marketed drugs. Therefore, the prediction of the binding affinity towards CYP2D6 and CYP2C9 would be beneficial for identifying cytochrome-mediated adverse effects triggered by drugs or chemicals (e.g., toxic reactions, drug-drug, and food-drug interactions). By identifying the binding mode by using pharmacophore prealignment, automated flexible docking, and by quantifying the binding affinity by multidimensional QSAR (mQSAR), we validated a model family of 56 compounds (46 training, 10 test) and 85 compounds (68 training, 17 test) for CYP2D6 and CYP2C9, respectively. The correlation with the experimental data (cross-validated r²=0.811 for CYP2D6 and 0.687 for CYP2C9) suggests that our approach is suited for predicting the binding affinity of compounds towards CYP2D6 and CYP2C9. The models were challenged by Y-scrambling and by testing an external dataset of binding compounds (15 compounds for CYP2D6 and 40 for CYP2C9). To assess the probability of false-positive predictions, datasets of nonbinders (64 compounds for CYP2D6 and 56 for CYP2C9) were tested by using the same protocol. The two validated mQSAR models were subsequently added to the VirtualToxLab (VTL, http://www.virtualtoxlab.org).
Collapse
Affiliation(s)
- Gianluca Rossato
- Institute of Molecular Pharmacy, Pharmacenter, University of Basel, Switzerland
| | | | | | | | | |
Collapse
|
33
|
Chemical inhibitors of cytochrome P450 isoforms in human liver microsomes: a re-evaluation of P450 isoform selectivity. Eur J Drug Metab Pharmacokinet 2011; 36:1-16. [PMID: 21336516 DOI: 10.1007/s13318-011-0024-2] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2010] [Accepted: 02/01/2011] [Indexed: 01/24/2023]
Abstract
The majority of marketed small-molecule drugs undergo metabolism by hepatic Cytochrome P450 (CYP) enzymes (Rendic 2002). Since these enzymes metabolize a structurally diverse number of drugs, metabolism-based drug-drug interactions (DDIs) can potentially occur when multiple drugs are coadministered to patients. Thus, a careful in vitro assessment of the contribution of various CYP isoforms to the total metabolism is important for predicting whether such DDIs might take place. One method of CYP phenotyping involves the use of potent and selective chemical inhibitors in human liver microsomal incubations in the presence of a test compound. The selectivity of such inhibitors plays a critical role in deciphering the involvement of specific CYP isoforms. Here, we review published data on the potency and selectivity of chemical inhibitors of the major human hepatic CYP isoforms. The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. As for CYP2A6, tranylcypromine is the most widely used inhibitor, but on the basis of initial studies, either 3-(pyridin-3-yl)-1H-pyrazol-5-yl)methanamine (PPM) and 3-(2-methyl-1H-imidazol-1-yl)pyridine (MIP) can replace tranylcypromine as the most selective CYP2A6 inhibitor. For CYP3A4, ketoconazole is widely used in phenotyping studies, although azamulin is a far more selective CYP3A inhibitor. Most of the phenotyping studies do not include CYP2E1, mostly because of the limited number of new drug candidates that are metabolized by this enzyme. Among the inhibitors for this enzyme, 4-methylpyrazole appears to be selective.
Collapse
|
34
|
Knych HKD, McKemie DS, Stanley SD. Molecular cloning, expression, and initial characterization of members of the CYP3A family in horses. Drug Metab Dispos 2010; 38:1820-7. [PMID: 20587621 DOI: 10.1124/dmd.110.032953] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The use of performance-enhancing drugs in the horse racing industry combined with the need for more rational approaches in the use of therapeutic agents in equids necessitates additional studies on the spectrum, content, and catalytic activities of hepatic cytochrome P450 monooxygenases in this species. In this study, three cytochrome P450 (P450) monooxygenases in the 3A family were cloned from, sequenced, and expressed in a baculovirus expression system. The proteins were designated CYP3A89, CYP3A96, and CYP3A97. Expression studies produced various results among the three proteins. CYP3A89 appears to undergo post-translational modification, producing a truncated protein, and although metabolically active, CYP3A97 did not have a detectable P450 spectrum. Expression of CYP3A96 produced a full-length, catalytically active protein. CYP3A96 catalyzed testosterone, and nifedipine metabolism was 20- and 10-fold slower, respectively, compared with the human counterpart, CYP3A4. Relative hepatic expression levels of each member of the CYP3A family, determined using quantitative reverse transcription-polymerase chain reaction, varied more than 1000-fold in individual horses. The results demonstrate substantial interspecies variability in metabolism of substrates by members of the CYP3A family in the horse and human and support the need to fully characterize 450-mediated metabolism in equids. These studies provide a framework for screening therapeutically useful drugs and provide a method for determination of metabolites of illegal performance-enhancing drugs without the time and expense of either in vivo studies or obtaining liver samples for in vitro analysis.
Collapse
Affiliation(s)
- Heather K DiMaio Knych
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California-Davis, West Health Science Drive, Davis, CA 95616, USA.
| | | | | |
Collapse
|
35
|
Parkinson A, Kazmi F, Buckley DB, Yerino P, Ogilvie BW, Paris BL. System-Dependent Outcomes during the Evaluation of Drug Candidates as Inhibitors of Cytochrome P450 (CYP) and Uridine Diphosphate Glucuronosyltransferase (UGT) Enzymes: Human Hepatocytes versus Liver Microsomes versus Recombinant Enzymes. Drug Metab Pharmacokinet 2010; 25:16-27. [DOI: 10.2133/dmpk.25.16] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
DiMaio Knych H, DeStefano Shields C, Buckpitt A, Stanley S. Equine cytochrome P450 2C92: cDNA cloning, expression and initial characterization. Arch Biochem Biophys 2009; 485:49-55. [DOI: 10.1016/j.abb.2009.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 02/11/2009] [Accepted: 02/12/2009] [Indexed: 12/12/2022]
|
37
|
Jones BC, Middleton DS, Youdim K. 6 Cytochrome P450 Metabolism and Inhibition: Analysis for Drug Discovery. PROGRESS IN MEDICINAL CHEMISTRY 2009; 47:239-63. [DOI: 10.1016/s0079-6468(08)00206-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
38
|
Complementary DNA cloning, functional expression and characterization of a novel cytochrome P450, CYP2D50, from equine liver. Biochem Pharmacol 2008; 76:904-11. [DOI: 10.1016/j.bcp.2008.07.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 07/04/2008] [Accepted: 07/11/2008] [Indexed: 11/21/2022]
|
39
|
Pelkonen O, Turpeinen M, Hakkola J, Honkakoski P, Hukkanen J, Raunio H. Inhibition and induction of human cytochrome P450 enzymes: current status. Arch Toxicol 2008; 82:667-715. [PMID: 18618097 DOI: 10.1007/s00204-008-0332-8] [Citation(s) in RCA: 386] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 06/16/2008] [Indexed: 02/07/2023]
Abstract
Variability of drug metabolism, especially that of the most important phase I enzymes or cytochrome P450 (CYP) enzymes, is an important complicating factor in many areas of pharmacology and toxicology, in drug development, preclinical toxicity studies, clinical trials, drug therapy, environmental exposures and risk assessment. These frequently enormous consequences in mind, predictive and pre-emptying measures have been a top priority in both pharmacology and toxicology. This means the development of predictive in vitro approaches. The sound prediction is always based on the firm background of basic research on the phenomena of inhibition and induction and their underlying mechanisms; consequently the description of these aspects is the purpose of this review. We cover both inhibition and induction of CYP enzymes, always keeping in mind the basic mechanisms on which to build predictive and preventive in vitro approaches. Just because validation is an essential part of any in vitro-in vivo extrapolation scenario, we cover also necessary in vivo research and findings in order to provide a proper view to justify in vitro approaches and observations.
Collapse
Affiliation(s)
- Olavi Pelkonen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, PO Box 5000 (Aapistie 5 B), 90014 Oulu, Finland.
| | | | | | | | | | | |
Collapse
|
40
|
Zhang H, Davis CD, Sinz MW, Rodrigues AD. Cytochrome P450 reaction-phenotyping: an industrial perspective. Expert Opin Drug Metab Toxicol 2008; 3:667-87. [PMID: 17916054 DOI: 10.1517/17425255.3.5.667] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It is now widely accepted that the fraction of the dose metabolized by a given drug-metabolizing enzyme is one of the major factors governing the magnitude of a drug interaction and the impact of a polymorphism on (total) drug clearance. Therefore, most pharmaceutical companies determine the enzymes involved in the metabolism of a new chemical entity (NCE) in vitro, in conjunction with human data on absorption, distribution, metabolism and excretion. This so called reaction-phenotyping, or isozyme-mapping, usually involves the use of multiple reagents (e.g., recombinant proteins, liver subcellular fractions, enzyme-selective chemical inhibitors and antibodies). For the human CYPs, reagents are readily available and in vitro reaction-phenotyping data are now routinely included in most regulatory documents. Ideally, the various metabolites have been definitively identified, incubation conditions have afforded robust kinetic analyses, and well characterized (high quality) reagents and human tissues have been employed. It is also important that the various in vitro data are consistent (e.g., scaled turnover with recombinant CYP proteins, CYP inhibition and correlation data with human liver microsomes) and enable an integrated in vitro CYP reaction-phenotype. Results of the in vitro CYP reaction-phenotyping are integrated with clinical data (e.g., human radiolabel and drug interaction studies) and a complete package is then submitted for regulatory review. If the NCE receives market approval, information on key routes of clearance and their associated potential for drug-drug interactions are included in the product label. The present review focuses on in vitro CYP reaction-phenotyping and the integration of data. Relatively simple strategies enabling the design and prioritization of follow up clinical studies are also discussed.
Collapse
Affiliation(s)
- Hongjian Zhang
- Bristol-Myers Squibb Research and Development, Pharmaceutical Candidate Optimization, PO Box 4000, Princeton, NJ 08543, USA.
| | | | | | | |
Collapse
|
41
|
Ghosal A, Ramanathan R, Yuan Y, Hapangama N, Chowdhury SK, Kishnani NS, Alton KB. Identification of human liver cytochrome P450 enzymes involved in biotransformation of vicriviroc, a CCR5 receptor antagonist. Drug Metab Dispos 2007; 35:2186-95. [PMID: 17827338 DOI: 10.1124/dmd.107.017517] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vicriviroc (SCH 417690), a CCR5 receptor antagonist, is currently under investigation for the treatment of human immunodeficiency virus infection. The objective of this study was to identify human liver cytochrome P450 enzyme(s) responsible for the metabolism of vicriviroc. Human liver microsomes metabolized vicriviroc via N-oxidation (M2/M3), O-demethylation (M15), N,N-dealkylation (M16), N-dealkylation (M41), and oxidation to a carboxylic acid metabolite (M35b/M37a). Recombinant human CYP3A4 catalyzed the formation of all these metabolites, whereas CYP3A5 catalyzed the formation of M2/M3 and M41. CYP2C9 only catalyzed the formation of M15. There was a high correlation between the rates of formation of M2/M3, M15, and M41, which was determined using 10 human liver microsomal samples and testosterone 6beta-hydroxylation catalyzed by CYP3A4/5 (r > or = 0.91). Ketoconazole and azamulin (inhibitors of CYP3A4) were potent inhibitors of the formation of M2/M3, M15, M41, and M35b/M37a from human liver microsomes. A CYP3A4/5-specific monoclonal antibody (1 microg/microg of protein) inhibited the formation of all metabolites from human liver microsomes by 86 to 100%. The results of this study suggest that formation of the major vicriviroc metabolites in human liver microsomes is primarily mediated via CYP3A4. CYP2C9 and CYP3A5 most likely play a minor role in the biotransformation of this compound. These enzymology data will provide guidance to design clinical studies to address any potential drug-drug interactions.
Collapse
Affiliation(s)
- Anima Ghosal
- Drug Metabolism and Pharmacokinetics, Schering-Plough Research Institute, Kenilworth, NJ 07033, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Wahlstrom JL, Rock DA, Slatter JG, Wienkers LC. Advances in predicting CYP-mediated drug interactions in the drug discovery setting. Expert Opin Drug Discov 2006; 1:677-91. [DOI: 10.1517/17460441.1.7.677] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|