1
|
Aurinsalo L, Lapatto‐Reiniluoto O, Kurkela M, Neuvonen M, Kiiski JI, Niemi M, Tornio A, Backman JT. A Phenotyping Tool for Seven Cytochrome P450 Enzymes and Two Transporters: Application to Examine the Effects of Clopidogrel and Gemfibrozil. Clin Pharmacol Ther 2025; 117:1732-1742. [PMID: 39982209 PMCID: PMC12087695 DOI: 10.1002/cpt.3610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Clinical cocktails for cytochrome P450 (CYP) phenotyping lack a marker for CYP2C8. We aimed to combine the CYP2C8 index drug repaglinide with the Geneva cocktail (caffeine/CYP1A2, bupropion/CYP2B6, flurbiprofen/CYP2C9, omeprazole/CYP2C19, dextromethorphan/CYP2D6, and midazolam/CYP3A4). We also included endogenous organic anion transporting polypeptide (OATP) 1B1 and 1B3 biomarkers glycochenodeoxycholate 3-O-glucuronide and glycochenodeoxycholate 3-sulfate, and investigated the CYP2C8 inhibition selectivity of clopidogrel and gemfibrozil with the full cocktail. In a five-phase randomized cross-over study, the following drugs were administered to 16 healthy volunteers: (i) repaglinide, (ii) the Geneva cocktail, (iii) repaglinide with the Geneva cocktail (full cocktail), (iv) clopidogrel followed by the full cocktail, and (v) gemfibrozil followed by the full cocktail. The Geneva cocktail increased repaglinide AUC0-23h 1.22-fold (90% confidence interval 1.04-1.44, P = 0.033). The full cocktail accurately captured known inhibitory effects of clopidogrel on CYP2B6, CYP2C8, and CYP2C19 and that of gemfibrozil on CYP2C8. Gemfibrozil decreased the paraxanthine/caffeine AUC0-12h ratio by 23% (14-31%, P < 0.01) and increased caffeine AUC0-12h 1.20-fold (1.03-1.40, P = 0.036). Gemfibrozil increased the metabolite-to-index drug AUC0-23h ratios of flurbiprofen, omeprazole, dextromethorphan, and midazolam 1.59-fold (1.32-1.92), 1.47-fold (1.34-1.61), 1.79-fold (1.23-2.59), and 2.1-fold (1.9-2.4), respectively, without affecting the index drug AUCs (P < 0.01). Gemfibrozil increased the AUC0-4h of glycochenodeoxycholate 3-O-glucuronide 1.33-fold (1.07-1.65, P = 0.027). In conclusion, the combination of repaglinide, the Geneva cocktail and endogenous biomarkers for OATP1B1 and OATP1B3 yields a nine-in-one phenotyping tool. Apart from strong CYP2C8 inhibition, gemfibrozil weakly inhibits CYP1A2 and OATP1B1 and appears to impair the elimination of the metabolites of several CYP index drugs.
Collapse
Affiliation(s)
- Laura Aurinsalo
- Department of Clinical PharmacologyUniversity of HelsinkiHelsinkiFinland
- Individualized Drug Therapy Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Clinical Pharmacology, HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
| | - Outi Lapatto‐Reiniluoto
- Department of Clinical PharmacologyUniversity of HelsinkiHelsinkiFinland
- Individualized Drug Therapy Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Clinical Pharmacology, HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
- HUS PharmacyHelsinki University HospitalHelsinkiFinland
| | - Mika Kurkela
- Department of Clinical PharmacologyUniversity of HelsinkiHelsinkiFinland
- Individualized Drug Therapy Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Mikko Neuvonen
- Department of Clinical PharmacologyUniversity of HelsinkiHelsinkiFinland
- Individualized Drug Therapy Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Johanna I. Kiiski
- Department of Clinical PharmacologyUniversity of HelsinkiHelsinkiFinland
- Individualized Drug Therapy Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Mikko Niemi
- Department of Clinical PharmacologyUniversity of HelsinkiHelsinkiFinland
- Individualized Drug Therapy Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Clinical Pharmacology, HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
| | - Aleksi Tornio
- Integrative Physiology and Pharmacology, Institute of BiomedicineUniversity of TurkuTurkuFinland
- Unit of Clinical PharmacologyTurku University HospitalTurkuFinland
| | - Janne T. Backman
- Department of Clinical PharmacologyUniversity of HelsinkiHelsinkiFinland
- Individualized Drug Therapy Research Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Department of Clinical Pharmacology, HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
| |
Collapse
|
2
|
Piha MOW, Cajanus K, Engström MT, Neuvonen M, Bergmann TK, Niemi M, Backman JT, Filppula AM, Tornio A. Candesartan Has No Clinically Meaningful Effect on the Plasma Concentrations of Cytochrome P450 2C8 Substrate Repaglinide in Humans. Drug Metab Dispos 2024; 52:1388-1395. [PMID: 39486868 DOI: 10.1124/dmd.124.001798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/07/2024] [Accepted: 10/03/2024] [Indexed: 11/04/2024] Open
Abstract
In vitro evidence shows that the acyl-β-D-glucuronide metabolite of candesartan inhibits cytochrome P450 (CYP) 2C8 with an inhibition constant of 7.12 μM. We investigated the effect of candesartan on the plasma concentrations and glucose-lowering effect of repaglinide, a sensitive clinical CYP2C8 index substrate. In a randomized crossover study, ten healthy volunteers ingested 8 mg of candesartan or placebo daily for three days, and on day 3, they also ingested 0.25 mg of repaglinide one hour after candesartan or placebo. We measured the plasma concentrations of repaglinide, candesartan, and candesartan acyl-β-D-glucuronide, and blood glucose concentrations for up to nine hours after repaglinide intake. Candesartan had no effect on the area under the plasma concentration-time curve and peak plasma concentration of repaglinide compared with placebo, with ratios of geometric means of 1.02 [P = 0.809; 90% confidence interval (CI) 0.90-1.15] and 1.13 (P = 0.346; 90% CI 0.90-1.43), respectively. Other pharmacokinetic variables and blood glucose concentrations were neither affected. Candesartan acyl-β-D-glucuronide was detectable in seven subjects, in whom the peak concentration of repaglinide was 1.32-fold higher in the candesartan phase than in the placebo phase (P = 0.041; 90% CI 1.07-1.62). Systemic concentrations of candesartan acyl-β-D-glucuronide were very low compared with its CYP2C8 inhibition constant (ratio ≪ 0.1). Furthermore, in a cohort of 93 cancer patients, no indication of decreased paclitaxel clearance was found in four patients using candesartan concomitantly. In conclusion, candesartan therapy is unlikely to inhibit CYP2C8-mediated metabolism of other drugs to any clinically significant extent. SIGNIFICANCE STATEMENT: The findings of this study suggest that candesartan is unlikely to cause drug-drug interactions via inhibition of cytochrome P450 (CYP) 2C8. Although candesartan acyl-β-D-glucuronide has been shown to inhibit CYP2C8 in vitro, it shows no clinically relevant CYP2C8 inhibition in humans due to low systemic concentrations.
Collapse
Affiliation(s)
- Mikael O W Piha
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Kristiina Cajanus
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Marica T Engström
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Mikko Neuvonen
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Troels K Bergmann
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Mikko Niemi
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Janne T Backman
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Anne M Filppula
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| | - Aleksi Tornio
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland (M.O.W.P., K.C., A.T.); Bioanalytical Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland (M.T.E.); Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland (M.O.W.P., K.C., A.T.); Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (M.Ne., M.Ni., J.T.B.); Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark and Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark (T.K.B.); Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni., J.T.B.); and Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland (A.M.F.)
| |
Collapse
|
3
|
Yu J, Rioux N, Gardner I, Owens K, Ragueneau-Majlessi I. Metabolite Measurement in Index Substrate Drug Interaction Studies: A Review of the Literature and Recent New Drug Application Reviews. Metabolites 2024; 14:522. [PMID: 39452902 PMCID: PMC11509402 DOI: 10.3390/metabo14100522] [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: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES Index substrates are used to understand the processes involved in pharmacokinetic (PK) drug-drug interactions (DDIs). The aim of this analysis is to review metabolite measurement in clinical DDI studies, focusing on index substrates for cytochrome P450 (CYP) enzymes, including CYP1A2 (caffeine), CYP2B6 (bupropion), CYP2C8 (repaglinide), CYP2C9 ((S)-warfarin, flurbiprofen), CYP2C19 (omeprazole), CYP2D6 (desipramine, dextromethorphan, nebivolol), and CYP3A (midazolam, triazolam). METHODS All data used in this evaluation were obtained from the Certara Drug Interaction Database. Clinical index substrate DDI studies with PK data for at least one metabolite, available from literature and recent new drug application reviews, were reviewed. Further, for positive DDI studies, a correlation analysis was performed between changes in plasma exposure of index substrates and their marker metabolites. RESULTS A total of 3261 individual index DDI studies were available, with 45% measuring at least one metabolite. The occurrence of metabolite measurement in clinical DDI studies varied widely between index substrates and enzymes. DISCUSSION AND CONCLUSIONS For substrates such as caffeine, bupropion, omeprazole, and dextromethorphan, the use of the metabolite/parent area under the curve ratio can provide greater sensitivity to DDI or reduce intrasubject variability. In some cases (e.g., omeprazole, repaglinide), the inclusion of metabolite measurement can provide mechanistic insights to understand complex interactions.
Collapse
Affiliation(s)
- Jingjing Yu
- Center of Excellence in Drug Interaction Science, Certara USA, 4 Radnor Corporate Center, Suite 350, Radnor, PA 19087, USA; (N.R.); (I.R.-M.)
| | - Nathalie Rioux
- Center of Excellence in Drug Interaction Science, Certara USA, 4 Radnor Corporate Center, Suite 350, Radnor, PA 19087, USA; (N.R.); (I.R.-M.)
| | - Iain Gardner
- Center of Excellence in Drug Interaction Science, Certara UK, 1 Concourse Way, Sheffield S1 2BJ, UK;
| | - Katie Owens
- Drug Interaction Solutions, Certara USA, 4 Radnor Corporate Center, Suite 350, Radnor, PA 19087, USA;
| | - Isabelle Ragueneau-Majlessi
- Center of Excellence in Drug Interaction Science, Certara USA, 4 Radnor Corporate Center, Suite 350, Radnor, PA 19087, USA; (N.R.); (I.R.-M.)
| |
Collapse
|
4
|
Di Paolo V, Ferrari FM, Veronese D, Poggesi I, Quintieri L. A genetic algorithm-based approach for the prediction of metabolic drug-drug interactions involving CYP2C8 or CYP2B6. J Pharmacol Toxicol Methods 2024; 127:107516. [PMID: 38777239 DOI: 10.1016/j.vascn.2024.107516] [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: 03/29/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND OBJECTIVES A genetic algorithm (GA) approach was developed to predict drug-drug interactions (DDIs) caused by cytochrome P450 2C8 (CYP2C8) inhibition or cytochrome P450 2B6 (CYP2B6) inhibition or induction. Nighty-eight DDIs, obtained from published in vivo studies in healthy volunteers, have been considered using the area under the plasma drug concentration-time curve (AUC) ratios (i.e., ratios of AUC of the drug substrate administered in combination with a DDI perpetrator to AUC of the drug substrate administered alone) to describe the extent of DDI. METHODS The following parameters were estimated in this approach: the contribution ratios (CRCYP2B6 and CRCYP2C8, i.e., the fraction of the dose metabolized via CYP2B6 or CYP2C8, respectively) and the inhibitory or inducing potency of the perpetrator drug (IRCYP2B6, IRCYP2C8 and ICCYP2B6, for inhibition of CYP2B6 and CYP2C8, and induction of CYP2B6, respectively). The workflow consisted of three main phases. First, the initial estimates of the parameters were estimated through GA. Then, the model was validated using an external validation. Finally, the parameter values were refined via a Bayesian orthogonal regression using all data. RESULTS The AUC ratios of 5 substrates, 11 inhibitors and 19 inducers of CYP2B6, and the AUC ratios of 19 substrates and 23 inhibitors of CYP2C8 were successfully predicted by the developed methodology within 50-200% of observed values. CONCLUSIONS The approach proposed in this work may represent a useful tool for evaluating the suitable doses of a CYP2C8 or CYP2B6 substrates co-administered with perpetrators.
Collapse
Affiliation(s)
- Veronica Di Paolo
- Laboratory of Drug Metabolism, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| | | | - Davide Veronese
- Laboratory of Drug Metabolism, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Italo Poggesi
- Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline S.p.A., Verona, Italy
| | - Luigi Quintieri
- Laboratory of Drug Metabolism, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
5
|
Tseng E, Lin J, Strelevitz TJ, DaSilva E, Goosen TC, Obach RS. Projections of Drug-Drug Interactions Caused by Time-Dependent Inhibitors of Cytochrome P450 1A2, 2B6, 2C8, 2C9, 2C19, and 2D6 Using In Vitro Data in Static and Dynamic Models. Drug Metab Dispos 2024; 52:DMD-AR-2024-001660. [PMID: 38408867 DOI: 10.1124/dmd.124.001660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
In vitro time-dependent inhibition (TDI) kinetic parameters for cytochrome P450 (CYP) 1A2, 2B6, 2C8, 2C9, 2C19, and 2D6, were determined in pooled human liver microsomes for 19 drugs (and 2 metabolites) for which clinical drug-drug interactions (DDI) are known. In vitro TDI data were incorporated into the projection of the magnitude of DDIs using mechanistic static models and Simcyp®. Results suggest that for the mechanistic static model, use of estimated average unbound exit concentration of the inhibitor from the liver resulted in a successful prediction of observed magnitude of clinical DDIs and was similar to Simcyp®. Overall, predictions of DDI magnitude (i.e., fold increase in AUC of a CYP-specific marker substrate) were within 2-fold of actual values. Geometric mean-fold errors were 1.7 and 1.6 for static and dynamic models, respectively. Projections of DDI from both models were also highly correlated to each other (r2 = 0.92). This investigation demonstrates that DDI can be reliably predicted from in vitro TDI data generated in HLM for several CYP enzymes. Simple mechanistic static model equations as well as more complex dynamic PBPK models can be employed in this process. Significance Statement Cytochrome P450 time-dependent inhibitors (TDI) can cause drug-drug interactions (DDI). An ability to reliably assess the potential for a new drug candidate to cause DDI is essential during drug development. In this report, TDI data for 19 drugs (and 2 metabolites) were measured and used in static and dynamic models to reliably project the magnitude of DDI resulting from inhibition of CYP1A2, 2B6, 2C8, 2C9, 2C19, and 2D6.
Collapse
Affiliation(s)
- Elaine Tseng
- Pfizer Global Research and Development, Pfizer Inc, United States
| | | | | | | | - Theunis C Goosen
- Pharmacokinetics, Dynamics & Metabolism, Pfizer, Inc, United States
| | | |
Collapse
|
6
|
Lai Y. The Role of Coproporphyrins As Endogenous Biomarkers for Organic Anion Transporting Polypeptide 1B Inhibition-Progress from 2016 to 2023. Drug Metab Dispos 2023; 51:950-961. [PMID: 37407093 DOI: 10.1124/dmd.122.001012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 07/07/2023] Open
Abstract
Since the initial clinical study investigating coproporphyrins I and III (CP-I and CP-III) as endogenous biomarkers for organic anion transporting polypeptide (OATP) inhibition drug-drug interactions (DDIs) published in 2016, significant progress has been made in confirming the usefulness of the CPs, particularly CP-I, as biomarkers in assessing OATP functions. CP-I exhibits selectivity toward OATP1B activity in human subjects with genetic variants of OATP1B1. Its sensitivity to a broad spectrum of clinical OATP1B inhibitors has been established from weak to vigorous. Dose-dependent CP-I changes in healthy human subjects show agreement with DDI magnitudes of probe substrates by rifampin treatment. Physiologically based pharmacokinetic models have been established for concentration changes of plasma CP-I with OATP inhibitors, demonstrating the usefulness of supporting the quantitative translation of the effect of CP-I levels into the DDI risk assessment of potential OATP inhibitors. As plasma CP-I's sensitivity, specificity, and selectivity have been validated in humans, monitoring CP-I levels in single and multiple clinical phase I dose escalation studies is recommended for early assessment of DDI risks and understanding the full dose-response of an investigational drug to OATP inhibitions. A decision tree is proposed to preclude the need to conduct a dedicated DDI study by administering a probe substrate drug to human subjects. SIGNIFICANCE STATEMENT: The minireview summarized the validation paths of coproporphyrins I and III (CP-I and CP-III) as biomarkers of organic anion transporting polypeptide 1B (OATP1B) inhibition in humans for their selectivity, specificity, and sensitivity. The utility of monitoring CP-I to assess drug-drug interactions of OATP1B inhibition in early drug development is proposed. Changes in plasma CP-I in phase I dose range studies can be used to frame plans for late-stage development and facilitate the mechanistic understanding of complex drug-drug interactions.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| |
Collapse
|
7
|
Kataoka M, Ohshiro S, Minami K, Hasegawa T, Higashino H, Takagi T, Togashi K, Mutaguchi K, Yamashita S. Contribution analysis of metabolic tissues on systemic exposure of an active metabolite after oral administration of verapamil using a stable isotope-labeled compound. Drug Metab Pharmacokinet 2023; 51:100514. [PMID: 37364522 DOI: 10.1016/j.dmpk.2023.100514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/18/2023] [Accepted: 05/11/2023] [Indexed: 06/28/2023]
Abstract
The present study illustrates the advantage of an isotope-IV study for the contribution analysis of metabolic tissues on systemic exposure of metabolites. A model parent drug, verapamil (VER), and its metabolite, norverapamil (Nor-VER), were used. This isotope-IV study used rats with and without the pre-treatment of the CYP inhibitor 1-aminobenzotriazole (ABT), was performed by the oral administration of VER (1 mg/kg) combined with the intravenous administration of stable isotope-labeled VER (VER-d6, 0.005 mg/kg). Plasma concentration profiles of both compounds and respective metabolites (Nor-VER, Nor-VER-d6) were then evaluated by LC-MSMS. VER oral availability was increased, and the systemic clearance decreased, in addition, the relative systemic exposure of Nor-VER and Nor-VER-d6 was increased by ABT pre-treatment. PK analyses revealed that, in ABT untreated rats, most Nor-VER in systemic circulation originated from the intestinal absorption process. ABT pre-treatment increased the contribution ratio to the systemic exposure of Nor-VER from the hepatic metabolism of systemically circulated VER, and decreased the contribution ratio of intestinal metabolism. These findings indicated that the isotope-IV study may be useful for considering the PK profile of metabolites.
Collapse
Affiliation(s)
- Makoto Kataoka
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan.
| | - Shota Ohshiro
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| | - Keiko Minami
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| | - Tsubasa Hasegawa
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| | - Haruki Higashino
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| | - Toshihide Takagi
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| | - Kazutaka Togashi
- Pharmaceutical Business Division, Sumika Chemical Analysis Service, Ltd., Osaka, 554-0022, Japan
| | - Kuninori Mutaguchi
- Pharmaceutical Business Division, Sumika Chemical Analysis Service, Ltd., Osaka, 554-0022, Japan
| | - Shinji Yamashita
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| |
Collapse
|
8
|
Bhatt S, Manhas D, Kumar V, Gour A, Sharma K, Dogra A, Ojha PK, Nandi U. Effect of Myricetin on CYP2C8 Inhibition to Assess the Likelihood of Drug Interaction Using In Silico, In Vitro, and In Vivo Approaches. ACS OMEGA 2022; 7:13260-13269. [PMID: 35474783 PMCID: PMC9026026 DOI: 10.1021/acsomega.2c00726] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/16/2022] [Indexed: 05/05/2023]
Abstract
Myricetin, a bioflavonoid, is widely used as functional food/complementary medicine and has promising multifaceted pharmacological actions against therapeutically validated anticancer targets. On the other hand, CYP2C8 is not only crucial for alteration in the pharmacokinetics of drugs to cause drug interaction but also unequivocally important for the metabolism of endogenous substances like the formation of epoxyeicosatrienoic acids (EETs), which are considered as signaling molecules against hallmarks of cancer. However, there is hardly any information known to date about the effect of myricetin on CYP2C8 inhibition and, subsequently, the CYP2C8-mediated drug interaction potential of myricetin at the preclinical/clinical level. We aimed here to explore the CYP2C8 inhibitory potential of myricetin using in silico, in vitro, and in vivo investigations. In the in vitro study, myricetin showed a substantial effect on CYP2C8 inhibition in human liver microsomes using CYP2C8-catalyzed amodiaquine-N-deethylation as an index reaction. Considering the Lineweaver-Burk plot, the Dixon plot, and the higher α-value, myricetin is found to be a mixed type of CYP2C8 inhibitor. Moreover, in vitro-in vivo extrapolation data suggest that myricetin is likely to cause drug interaction at the hepatic level. The molecular docking study depicted a strong interaction between myricetin and the active site of the human CYP2C8 enzyme. Moreover, myricetin caused considerable elevation in the oral exposure of amodiaquine as a CYP2C8 substrate via a slowdown of amodiaquine clearance in the rat model. Overall, the potent action of myricetin on CYP2C8 inhibition indicates that there is a need for further exploration to avoid drug interaction-mediated precipitation of obvious adverse effects as well as to optimize anticancer therapy.
Collapse
Affiliation(s)
- Shipra Bhatt
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Diksha Manhas
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vinay Kumar
- Drug
Theoretics and Chemoinformatics Laboratory, Department of Pharmaceutical
Technology, Jadavpur University, Kolkata 700032, India
| | - Abhishek Gour
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuhu Sharma
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
| | - Ashish Dogra
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Probir Kumar Ojha
- Drug
Theoretics and Chemoinformatics Laboratory, Department of Pharmaceutical
Technology, Jadavpur University, Kolkata 700032, India
| | - Utpal Nandi
- PK-PD
Toxicology (PPT) Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- ,
| |
Collapse
|
9
|
Di Paolo V, Ferrari FM, Poggesi I, Quintieri L. A Quantitative Approach to the Prediction of Drug-Drug Interactions Mediated by Cytochrome P450 2C8 Inhibition. Expert Opin Drug Metab Toxicol 2021; 17:1345-1352. [PMID: 34720033 DOI: 10.1080/17425255.2021.1998453] [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] [Indexed: 10/19/2022]
Abstract
BACKGROUND Ohno and Colleagues proposed an approach for predicting drug-drug interactions (DDIs) mediated by cytochrome P450 (CYP) 3A4 based on the use of the ratio of the inhibited to non-inhibited area under the plasma concentration time curve (AUC) of substrates to estimate the fraction of the dose metabolized via CYP3A4 (contribution ratio, CR) and the in vivo inhibitory potency of a perpetrator (inhibition ratio, IR). This study evaluated the performance of this approach on DDIs mediated by CYP2C8 inhibitors. RESEARCH DESIGN AND METHODS Initial estimates of CR and IR of CYP2C8 substrates and inhibitors were calculated for 33 DDI in vivo studies. The approach was externally validated with 17 additional studies. Bayesian orthogonal regression was used to refine the estimates of the parameters. Assessment of prediction success was conducted by plotting observed versus predicted AUC ratios. RESULTS Final estimates of CRs and IRs were obtained for 19 CYP2C8 substrates and 23 inhibitors, respectively. The method demonstrated good predictive capacity, with only two values outside of the prespecified limits. CONCLUSIONS The approach may help to adapt dose regimens for CYP2C8 substrates when given in combination with CYP2C8 inhibitors and to map the potential DDIs of new molecular entities.
Collapse
Affiliation(s)
- Veronica Di Paolo
- Laboratory of Drug Metabolism, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Italo Poggesi
- Department Clinical Pharmacology and Pharmacometrics, Janssen-Cilag S.p.A, Cologno Monzese, Italy
| | - Luigi Quintieri
- Laboratory of Drug Metabolism, Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
10
|
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
|
11
|
Dai X, Karol MD, Hitron M, Hard ML, Goulet MT, McLaughlin CF, Brantley SJ. Napabucasin Drug-Drug Interaction Potential, Safety, Tolerability, and Pharmacokinetics Following Oral Dosing in Healthy Adult Volunteers. Clin Pharmacol Drug Dev 2021; 10:824-839. [PMID: 34107166 PMCID: PMC8453567 DOI: 10.1002/cpdd.961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 04/10/2021] [Indexed: 12/05/2022]
Abstract
Napabucasin is an orally administered reactive oxygen species generator that is bioactivated by the intracellular antioxidant nicotinamide adenine dinucleotide phosphate:quinone oxidoreductase 1. Napabucasin induces cell death in cancer cells, including cancer stem cells. This phase 1 study (NCT03411122) evaluated napabucasin drug‐drug interaction potential for 7 cytochrome P450 (CYP) enzymes and the breast cancer resistance protein transporter/organic anion transporter 3. Healthy volunteers who tolerated napabucasin during period 1 received probe drugs during period 2, and in period 3 received napabucasin (240 mg twice daily; days 1‐11) plus a phenotyping cocktail containing omeprazole (CYP2C19), caffeine (CYP1A2), flurbiprofen (CYP2C9), bupropion (CYP2B6), dextromethorphan (CYP2D6), midazolam (CYP3A) (all oral; day 6), intravenous midazolam (day 7), repaglinide (CYP2C8; day 8), and rosuvastatin (breast cancer resistance protein/organic anion transporter 3; day 9). Drug‐drug interaction potential was evaluated in 17 of 30 enrolled volunteers. Napabucasin coadministration increased the area under the plasma concentration–time curve from time 0 extrapolated to infinity (geometric mean ratio [90% confidence interval]) of caffeine (124% [109.0%‐141.4%]), intravenous midazolam (118% [94.4%‐147.3%]), repaglinide (127% [104.7%‐153.3%]), and rosuvastatin (213% [42.5%‐1068.3%]) and decreased the area under the plasma concentration–time curve from time 0 extrapolated to infinity of dextromethorphan (71% [47.1%‐108.3%]), bupropion (79% [64.6%‐97.0%]), and hydroxybupropion (45% [15.7%‐129.6%]). No serious adverse events/deaths were reported. Generally, napabucasin is not expected to induce/inhibit drug clearance to a clinically meaningful degree.
Collapse
Affiliation(s)
- Xiaoshu Dai
- Clinical Pharmacology and Drug Metabolism, Sumitomo Dainippon Pharma Oncology, Inc., Cambridge, Massachusetts, USA
| | - Michael D Karol
- Clinical Pharmacology and Drug Metabolism, Sumitomo Dainippon Pharma Oncology, Inc., Cambridge, Massachusetts, USA
| | - Matthew Hitron
- Clinical Development, Sumitomo Dainippon Pharma Oncology, Inc., Cambridge, Massachusetts, USA
| | - Marjie L Hard
- Clinical Pharmacology, Nuventra, Inc., Durham, North Carolina, USA.,Current address: Praxis Precision Medicines, Cambridge, Massachusetts, USA
| | - Matthew T Goulet
- Clinical Pharmacology, Nuventra, Inc., Durham, North Carolina, USA.,Current address: Program Management Department, KSQ Therapeutics, Cambridge, Massachusetts, USA
| | | | - Scott J Brantley
- Clinical NCA & PK/PD, Nuventra, Inc., Durham, North Carolina, USA
| |
Collapse
|
12
|
Katayama N, Odagiri K, Hakamata A, Kamiya C, Uchida S, Tanaka S, Inui N, Namiki N, Tatsumi K, Watanabe H. Clinical evaluation of drug-drug interactions between the cytochrome P450 substrates selexipag and clopidogrel in Japanese volunteers. Br J Clin Pharmacol 2020; 87:1903-1911. [PMID: 32997809 DOI: 10.1111/bcp.14579] [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: 04/09/2020] [Revised: 09/07/2020] [Accepted: 09/12/2020] [Indexed: 11/30/2022] Open
Abstract
AIMS The strong cytochrome P450 (CYP) 2C8 inhibitor gemfibrozil has been demonstrated to increase the area under the plasma concentration-time curve from 0 to infinity (AUC0-∞ ) of ACT-333679, an active metabolite of selexipag, by 11-fold. Similarly to gemfibrozil, the CYP2C8 inhibitor clopidogrel increased ACT-333679 concentration by 1.9-fold after a single loading dose (300 mg once daily) and 2.7-fold after repeated treatment with the maintenance dose (75 mg once daily) in Europeans. However, the effects of clopidogrel on the pharmacokinetics of selexipag and ACT-333679 have not been fully elucidated in the Japanese population. METHODS We investigated the effect of clopidogrel on the pharmacokinetics of selexipag and ACT-333679 in 14 healthy Japanese volunteers. RESULTS The concomitant administration of clopidogrel with selexipag did not influence the maximum concentration and AUC0-∞ of selexipag, whereas it significantly increased AUC0-∞ of ACT-333679 by approximately 1.90-fold (90% confidence interval 1.69-2.14) without changing the maximum concentration. When selexipag was administered 1 day after clopidogrel was discontinued, the increase in AUC0-∞ of ACT-333679 was 1.37-fold (90% confidence interval 0.93-2.02), suggesting that, although the inhibitory effect of clopidogrel on CYP2C8 was reduced, it persisted for at least 1 day after withdrawal. CONCLUSION Our results demonstrated the impact of clopidogrel on the pharmacokinetics of selexipag and its active metabolite and suggested that selexipag should be carefully prescribed with clopidogrel with dose adjustment or reducing the dosing frequency in Japanese clinical settings.
Collapse
Affiliation(s)
- Naoki Katayama
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Akio Hakamata
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Chiaki Kamiya
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinya Uchida
- Department of Pharmacy Practice and Science, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Shimako Tanaka
- Department of Pharmacy Practice and Science, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Naoki Inui
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Namiki
- Department of Pharmacy Practice and Science, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroshi Watanabe
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
13
|
Türk D, Hanke N, Wolf S, Frechen S, Eissing T, Wendl T, Schwab M, Lehr T. Physiologically Based Pharmacokinetic Models for Prediction of Complex CYP2C8 and OATP1B1 (SLCO1B1) Drug-Drug-Gene Interactions: A Modeling Network of Gemfibrozil, Repaglinide, Pioglitazone, Rifampicin, Clarithromycin and Itraconazole. Clin Pharmacokinet 2020; 58:1595-1607. [PMID: 31129789 PMCID: PMC6885506 DOI: 10.1007/s40262-019-00777-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Drug–drug interactions (DDIs) and drug–gene interactions (DGIs) pose a serious health risk that can be avoided by dose adaptation. These interactions are investigated in strictly controlled setups, quantifying the effect of one perpetrator drug or polymorphism at a time, but in real life patients frequently take more than two medications and are very heterogenous regarding their genetic background. Objectives The first objective of this study was to provide whole-body physiologically based pharmacokinetic (PBPK) models of important cytochrome P450 (CYP) 2C8 perpetrator and victim drugs, built and evaluated for DDI and DGI studies. The second objective was to apply these models to describe complex interactions with more than two interacting partners. Methods PBPK models of the CYP2C8 and organic-anion-transporting polypeptide (OATP) 1B1 perpetrator drug gemfibrozil (parent–metabolite model) and the CYP2C8 victim drugs repaglinide (also an OATP1B1 substrate) and pioglitazone were developed using a total of 103 clinical studies. For evaluation, these models were applied to predict 34 different DDI studies, establishing a CYP2C8 and OATP1B1 PBPK DDI modeling network. Results The newly developed models show a good performance, accurately describing plasma concentration–time profiles, area under the plasma concentration–time curve (AUC) and maximum plasma concentration (Cmax) values, DDI studies as well as DGI studies. All 34 of the modeled DDI AUC ratios (AUC during DDI/AUC control) and DDI Cmax ratios (Cmax during DDI/Cmax control) are within twofold of the observed values. Conclusions Whole-body PBPK models of gemfibrozil, repaglinide, and pioglitazone have been built and qualified for DDI and DGI prediction. PBPK modeling is applicable to investigate complex interactions between multiple drugs and genetic polymorphisms. Electronic supplementary material The online version of this article (10.1007/s40262-019-00777-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Denise Türk
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany
| | - Nina Hanke
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany
| | - Sarah Wolf
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany
| | | | | | - Thomas Wendl
- Clinical Pharmacometrics, Bayer AG, Leverkusen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
- Department of Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany.
| |
Collapse
|
14
|
Kimoto E, Obach RS, Varma MV. Identification and quantitation of enzyme and transporter contributions to hepatic clearance for the assessment of potential drug-drug interactions. Drug Metab Pharmacokinet 2020; 35:18-29. [DOI: 10.1016/j.dmpk.2019.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/30/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022]
|
15
|
Liu X. Transporter-Mediated Drug-Drug Interactions and Their Significance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:241-291. [PMID: 31571167 DOI: 10.1007/978-981-13-7647-4_5] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug transporters are considered to be determinants of drug disposition and effects/toxicities by affecting the absorption, distribution, and excretion of drugs. Drug transporters are generally divided into solute carrier (SLC) family and ATP binding cassette (ABC) family. Widely studied ABC family transporters include P-glycoprotein (P-GP), breast cancer resistance protein (BCRP), and multidrug resistance proteins (MRPs). SLC family transporters related to drug transport mainly include organic anion-transporting polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), organic cation/carnitine transporters (OCTNs), peptide transporters (PEPTs), and multidrug/toxin extrusions (MATEs). These transporters are often expressed in tissues related to drug disposition, such as the small intestine, liver, and kidney, implicating intestinal absorption of drugs, uptake of drugs into hepatocytes, and renal/bile excretion of drugs. Most of therapeutic drugs are their substrates or inhibitors. When they are comedicated, serious drug-drug interactions (DDIs) may occur due to alterations in intestinal absorption, hepatic uptake, or renal/bile secretion of drugs, leading to enhancement of their activities or toxicities or therapeutic failure. This chapter will illustrate transporter-mediated DDIs (including food drug interaction) in human and their clinical significances.
Collapse
Affiliation(s)
- Xiaodong Liu
- China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
16
|
Giri P, Delvadia P, Ladani MK, Prajapati N, Gupta L, Patel N, Joshi V, Giri S, Jain MR, Srinivas NR, Patel PR. Lack of inhibition of CYP2C8 by saroglitazar magnesium: In vivo assessment using montelukast, rosiglitazone, pioglitazone, repaglinide and paclitaxel as victim drugs in Wistar rats. Eur J Pharm Sci 2019; 130:107-113. [PMID: 30633968 DOI: 10.1016/j.ejps.2019.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/01/2018] [Accepted: 01/08/2019] [Indexed: 12/22/2022]
Abstract
Saroglitazar, a PPAR αҮ agonist, is currently undergoing global development for the treatment of NASH and other indications. Saroglitazar showed CYP2C8 inhibition in human liver microsomes (IC50: 2.9 μM). The aim was to carry out drug-drug interaction (DDI) studies in Wistar rats using saroglitazar (perpetrator drug) with five CYP2C8 substrates. Also, the in vitro CYP2C8 inhibitory potential of saroglitazar in rat liver microsomes (RLM) was evaluated to justify use of preclinical model. The oral pharmacokinetics of various CYP2C8 substrates; montelukast, rosiglitazone, pioglitazone, repaglinide and intravenous pharmacokinetics of paclitaxel was assessed in the presence/absence of oral saroglitazar (4 mg/kg) in Wistar rats. A separate study was performed to assess the oral pharmacokinetics of saroglitazar. Serial blood samples were collected from all studies and the harvested plasma were stored frozen until bioanalysis. LC-MS/MS was used for the analysis of various analytes; concentration data was subjected to noncompartmental pharmacokinetic analysis. Statistical tests (unpaired t-test) were employed to judge the level of DDI. Generally, the pharmacokinetics of CYP2C8 substrates was not affected by the concomitant intake of saroglitazar as judged by the overall exposure (AUC0-last and AUC0-inf) and elimination half-life. The CYP2C8 IC50 of 4.5 μM in RLM for saroglitazar, supported the use of rats for this DDI study. In conclusion, pharmacokinetic data of diverse CYP2C8 substrates suggested that coadministration of saroglitazar did not cause clinically relevant DDI.
Collapse
Affiliation(s)
- Poonam Giri
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India.
| | - Prashant Delvadia
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Meera K Ladani
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Namrata Prajapati
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Lakshmikant Gupta
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Nirmal Patel
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Vipul Joshi
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Shyamkumar Giri
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Mukul R Jain
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India; Department of Pharmacology, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Nuggehally R Srinivas
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Moraiya, Ahmadabad, Gujarat, India
| | - Pankaj R Patel
- Zydus Research Centre, Cadila Healthcare Ltd., Satellite cross road, Ahmadabad, Gujarat, India
| |
Collapse
|
17
|
Tan ML, Zhao P, Zhang L, Ho YF, Varma MVS, Neuhoff S, Nolin TD, Galetin A, Huang SM. Use of Physiologically Based Pharmacokinetic Modeling to Evaluate the Effect of Chronic Kidney Disease on the Disposition of Hepatic CYP2C8 and OATP1B Drug Substrates. Clin Pharmacol Ther 2018; 105:719-729. [PMID: 30074626 PMCID: PMC8246729 DOI: 10.1002/cpt.1205] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/30/2018] [Indexed: 12/15/2022]
Abstract
Chronic kidney disease (CKD) differentially affects the pharmacokinetics (PK) of nonrenally cleared drugs via certain pathways (e.g., cytochrome P450 (CYP)2D6); however, the effect on CYP2C8‐mediated clearance is not well understood because of overlapping substrate specificity with hepatic organic anion‐transporting polypeptides (OATPs). This study used physiologically based pharmacokinetic (PBPK) modeling to delineate potential changes in CYP2C8 or OATP1B activity in patients with CKD. Drugs analyzed are predominantly substrates of CYP2C8 (rosiglitazone and pioglitazone), OATP1B (pitavastatin), or both (repaglinide). Following initial model verification, pharmacokinetics (PK) of these drugs were simulated in patients with severe CKD considering changes in glomerular filtration rate (GFR), plasma protein binding, and activity of either CYP2C8 and/or OATP1B in a stepwise manner. The PBPK analysis suggests that OATP1B activity could be decreased up to 60% in severe CKD, whereas changes to CYP2C8 are negligible. This improved understanding of CKD effect on clearance pathways could be important to inform the optimal use of nonrenally eliminated drugs in patients with CKD.
Collapse
Affiliation(s)
- Ming-Liang Tan
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ping Zhao
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.,Quantitative Sciences, Global Health-Integrated Development, Bill and Melinda Gates Foundation, Seattle, Washington, USA
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.,Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yunn-Fang Ho
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Manthena V S Varma
- Pharmacokinetics, Pharmacodynamics & Metabolism Department-New Chemical Entities, Pfizer Inc., Groton, Connecticut, USA
| | | | - Thomas D Nolin
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, and Department of Medicine Renal-Electrolyte Division, Schools of Pharmacy and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Heath Sciences, University of Manchester, Manchester, UK
| | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
18
|
Alam K, Crowe A, Wang X, Zhang P, Ding K, Li L, Yue W. Regulation of Organic Anion Transporting Polypeptides (OATP) 1B1- and OATP1B3-Mediated Transport: An Updated Review in the Context of OATP-Mediated Drug-Drug Interactions. Int J Mol Sci 2018. [PMID: 29538325 PMCID: PMC5877716 DOI: 10.3390/ijms19030855] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Organic anion transporting polypeptides (OATP) 1B1 and OATP1B3 are important hepatic transporters that mediate the uptake of many clinically important drugs, including statins from the blood into the liver. Reduced transport function of OATP1B1 and OATP1B3 can lead to clinically relevant drug-drug interactions (DDIs). Considering the importance of OATP1B1 and OATP1B3 in hepatic drug disposition, substantial efforts have been given on evaluating OATP1B1/1B3-mediated DDIs in order to avoid unwanted adverse effects of drugs that are OATP substrates due to their altered pharmacokinetics. Growing evidences suggest that the transport function of OATP1B1 and OATP1B3 can be regulated at various levels such as genetic variation, transcriptional and post-translational regulation. The present review summarizes the up to date information on the regulation of OATP1B1 and OATP1B3 transport function at different levels with a focus on potential impact on OATP-mediated DDIs.
Collapse
Affiliation(s)
- Khondoker Alam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Alexandra Crowe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Xueying Wang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Pengyue Zhang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kai Ding
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA.
| | - Lang Li
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA.
| | - Wei Yue
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| |
Collapse
|
19
|
Pakkir Maideen NM, Manavalan G, Balasubramanian K. Drug interactions of meglitinide antidiabetics involving CYP enzymes and OATP1B1 transporter. Ther Adv Endocrinol Metab 2018; 9:259-268. [PMID: 30181852 PMCID: PMC6116761 DOI: 10.1177/2042018818767220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/16/2018] [Indexed: 12/11/2022] Open
Abstract
Meglitinides such as repaglinide and nateglinide are useful to treat type 2 diabetes patients who follow a flexible lifestyle. They are short-acting insulin secretagogues and are associated with less risk of hypoglycemia, weight gain and chronic hyperinsulinemia compared with sulfonylureas. Meglitinides are the substrates of cytochrome P450 (CYP) enzymes and organic anion transporting polypeptide 1B1 (OATP1B1 transporter) and the coadministration of the drugs affecting them will result in pharmacokinetic drug interactions. This article focuses on the drug interactions of meglitinides involving CYP enzymes and OATP1B1 transporter. To prevent the risk of hypoglycemic episodes, prescribers and pharmacists must be aware of the adverse drug interactions of meglitinides.
Collapse
|
20
|
Cleary Y, Gertz M, Morcos PN, Yu L, Youdim K, Phipps A, Fowler S, Parrott N. Model-Based Assessments of CYP-Mediated Drug-Drug Interaction Risk of Alectinib: Physiologically Based Pharmacokinetic Modeling Supported Clinical Development. Clin Pharmacol Ther 2017; 104:505-514. [DOI: 10.1002/cpt.956] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/08/2017] [Accepted: 11/22/2017] [Indexed: 12/15/2022]
Affiliation(s)
| | | | | | - Li Yu
- Roche Innovation Center; New York New York USA
| | | | | | | | | |
Collapse
|
21
|
Shebley M, Fu W, Badri P, Bow DAJ, Fischer V. Physiologically Based Pharmacokinetic Modeling Suggests Limited Drug-Drug Interaction Between Clopidogrel and Dasabuvir. Clin Pharmacol Ther 2017; 102:679-687. [PMID: 28411400 PMCID: PMC5599937 DOI: 10.1002/cpt.689] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 03/10/2017] [Accepted: 03/11/2017] [Indexed: 12/14/2022]
Abstract
Dasabuvir, a nonnucleoside NS5B polymerase inhibitor, is a sensitive substrate of cytochrome P450 (CYP) 2C8 with a potential for drug-drug interaction (DDI) with clopidogrel. A physiologically based pharmacokinetic (PBPK) model was developed for dasabuvir to evaluate the DDI potential with clopidogrel, the acyl-β-D glucuronide metabolite of which has been reported as a strong mechanism-based inhibitor of CYP2C8 based on an interaction with repaglinide. In addition, the PBPK model for clopidogrel and its metabolite were updated with additional in vitro data. Sensitivity analyses using these PBPK models suggested that CYP2C8 inhibition by clopidogrel acyl-β-D glucuronide may not be as potent as previously suggested. The dasabuvir and updated clopidogrel PBPK models predict a moderate increase of 1.5-1.9-fold for Cmax and 1.9-2.8-fold for AUC of dasabuvir when coadministered with clopidogrel. While the PBPK results suggest there is a potential for DDI between dasabuvir and clopidogrel, the magnitude is not expected to be clinically relevant.
Collapse
Affiliation(s)
- M Shebley
- Drug Metabolism, Pharmacokinetics and BioanalysisAbbVie Inc.North ChicagoIllinoisUSA
- Clinical Pharmacology and PharmacometricsAbbVie Inc.North ChicagoIllinoisUSA
| | - W Fu
- Drug Metabolism, Pharmacokinetics and BioanalysisAbbVie Inc.North ChicagoIllinoisUSA
- U.S. Food and Drug Administration, CDEROffice of Clinical PharmacologySilver SpringMarylandUSA
| | - P Badri
- Clinical Pharmacology and PharmacometricsAbbVie Inc.North ChicagoIllinoisUSA
- Vertex PharmaceuticalsBostonMassachusettsUSA
| | - DAJ Bow
- Drug Metabolism, Pharmacokinetics and BioanalysisAbbVie Inc.North ChicagoIllinoisUSA
| | - V Fischer
- Drug Metabolism, Pharmacokinetics and BioanalysisAbbVie Inc.North ChicagoIllinoisUSA
| |
Collapse
|
22
|
Haraya K, Kato M, Chiba K, Sugiyama Y. Prediction of inter-individual variability on the pharmacokinetics of CYP2C8 substrates in human. Drug Metab Pharmacokinet 2017; 32:277-285. [PMID: 29174535 DOI: 10.1016/j.dmpk.2017.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/06/2017] [Accepted: 09/06/2017] [Indexed: 01/10/2023]
Abstract
Inter-individual variability in pharmacokinetics can lead to unexpected side effects and treatment failure, and is therefore an important factor in drug development. CYP2C8 is a major drug-metabolizing enzyme known to be involved in the metabolism of over 100 drugs. In this study, we predicted the inter-individual variability in AUC/Dose of CYP2C8 substrates in healthy volunteers using the Monte Carlo simulation. Inter-individual variability in the hepatic intrinsic clearance of CYP2C8 substrates (CLint,h,2C8) was estimated from the inter-individual variability in pharmacokinetics of pioglitazone, which is a major CYP2C8 substrate. The coefficient of variation (CV) of CLint,h,2C8 was estimated to be 40%. Using this value, the CVs of AUC/Dose of other major CYP2C8 substrates, rosiglitazone and amodiaquine, were predicted to validate the estimated CV of CLint,h,2C8. As a result, the reported CVs of both substrates were within the 2.5-97.5 percentile range of the predicted CVs. Furthermore, the CVs of AUC/Dose of the CYP2C8 substrates loperamide and chloroquine, which are affected by renal clearance, were also successfully predicted. Combining this value with previously reported CVs of other CYPs, we were able to successfully predict the inter-individual variability in pharmacokinetics of various drugs in clinical.
Collapse
Affiliation(s)
- Kenta Haraya
- Chugai Pharmabody Research Pte. Ltd., Singapore.
| | | | - Koji Chiba
- Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Yokohama, Japan; Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Yokohama, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Yokohama, Japan
| |
Collapse
|
23
|
Quantitative Analyses of the Influence of Parameters Governing Rate-Determining Process of Hepatic Elimination of Drugs on the Magnitudes of Drug-Drug Interactions via Hepatic OATPs and CYP3A Using Physiologically Based Pharmacokinetic Models. J Pharm Sci 2017; 106:2739-2750. [DOI: 10.1016/j.xphs.2017.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/20/2017] [Accepted: 05/01/2017] [Indexed: 01/20/2023]
|
24
|
Ma Y, Fu Y, Khojasteh SC, Dalvie D, Zhang D. Glucuronides as Potential Anionic Substrates of Human Cytochrome P450 2C8 (CYP2C8). J Med Chem 2017; 60:8691-8705. [DOI: 10.1021/acs.jmedchem.7b00510] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | | | | | - Deepak Dalvie
- Celgene Corporation, 10300 Campus
Point Drive, San Diego California 92121, United States
| | | |
Collapse
|
25
|
Stage TB, Bergmann TK, Kroetz DL. Clinical Pharmacokinetics of Paclitaxel Monotherapy: An Updated Literature Review. Clin Pharmacokinet 2017; 57:7-19. [DOI: 10.1007/s40262-017-0563-z] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
26
|
Yoshikado T, Maeda K, Furihata S, Terashima H, Nakayama T, Ishigame K, Tsunemoto K, Kusuhara H, Furihata KI, Sugiyama Y. A Clinical Cassette Dosing Study for Evaluating the Contribution of Hepatic OATPs and CYP3A to Drug-Drug Interactions. Pharm Res 2017; 34:1570-1583. [DOI: 10.1007/s11095-017-2168-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/27/2017] [Indexed: 10/19/2022]
|
27
|
Johansson S, Rosenbaum DP, Ahlqvist M, Rollison H, Knutsson M, Stefansson B, Elebring M. Effects of Tenapanor on Cytochrome P450-Mediated Drug-Drug Interactions. Clin Pharmacol Drug Dev 2017; 6:466-475. [PMID: 28301096 PMCID: PMC5599994 DOI: 10.1002/cpdd.346] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/26/2017] [Indexed: 01/15/2023]
Abstract
Tenapanor (RDX5791, AZD1722) is an inhibitor of sodium/hydrogen exchanger isoform 3 in development for the treatment of constipation‐predominant irritable bowel syndrome and the treatment of hyperphosphatemia in patients with chronic kidney disease on dialysis. We aimed to investigate whether tenapanor inhibits or induces cytochrome P450s (CYPs). In vitro experiments assessing the potential of tenapanor to affect various CYPs indicated that it could inhibit CYP3A4/5 (IC50 0.4‐0.7 μM). An open‐label, phase 1 clinical study (NCT02140268) evaluated the pharmacokinetics of the CYP3A4 substrate midazolam when administered with and without tenapanor. Healthy volunteers received a single oral dose of midazolam 7.5 mg on day 1 followed by tenapanor 15 mg twice daily on days 2 to 15, with an additional single 7.5‐mg midazolam dose coadministered on day 15. Midazolam exposure was similar whether it was administered alone or with tenapanor (geometric least‐squares mean ratio [90%CI] for [midazolam + tenapanor]/midazolam: area under the concentration‐time curve, 107% [101% to 113%]; Cmax 104% [89.6% to 122%]). Findings were similar for metabolites of midazolam. These results indicate that tenapanor 15 mg twice daily does not have a clinically relevant impact on CYP3A4 in humans and suggest that tenapanor can be coadministered with CYP3A4‐metabolized drugs without affecting their exposure.
Collapse
|
28
|
Fowler S, Morcos PN, Cleary Y, Martin-Facklam M, Parrott N, Gertz M, Yu L. Progress in Prediction and Interpretation of Clinically Relevant Metabolic Drug-Drug Interactions: a Minireview Illustrating Recent Developments and Current Opportunities. CURRENT PHARMACOLOGY REPORTS 2017; 3:36-49. [PMID: 28261547 PMCID: PMC5315728 DOI: 10.1007/s40495-017-0082-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW This review gives a perspective on the current "state of the art" in metabolic drug-drug interaction (DDI) prediction. We highlight areas of successful prediction and illustrate progress in areas where limits in scientific knowledge or technologies prevent us from having full confidence. RECENT FINDINGS Several examples of success are highlighted. Work done for bitopertin shows how in vitro and clinical data can be integrated to give a model-based understanding of pharmacokinetics and drug interactions. The use of interpolative predictions to derive explicit dosage recommendations for untested DDIs is discussed using the example of ibrutinib, and the use of DDI predictions in lieu of clinical studies in new drug application packages is exemplified with eliglustat and alectinib. Alectinib is also an interesting case where dose adjustment is unnecessary as the activity of a major metabolite compensates sufficiently for changes in parent drug exposure. Examples where "unusual" cytochrome P450 (CYP) and non-CYP enzymes are responsible for metabolic clearance have shown the importance of continuing to develop our repertoire of in vitro regents and techniques. The time-dependent inhibition assay using human hepatocytes suspended in full plasma allowed improved DDI predictions, illustrating the importance of continued in vitro assay development and refinement. SUMMARY During the past 10 years, a highly mechanistic understanding has been developed in the area of CYP-mediated metabolic DDIs enabling the prediction of clinical outcome based on preclinical studies. The combination of good quality in vitro data and physiologically based pharmacokinetic modeling may now be used to evaluate DDI risk prospectively and are increasingly accepted in lieu of dedicated clinical studies.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Peter N. Morcos
- Pharmaceutical Reseach and Early Development, Roche Innovation Center New York, F. Hoffmann-La Roche Ltd., 430 East 29th Street, New York City, NY USA
| | - Yumi Cleary
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Meret Martin-Facklam
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Neil Parrott
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Michael Gertz
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Li Yu
- Pharmaceutical Reseach and Early Development, Roche Innovation Center New York, F. Hoffmann-La Roche Ltd., 430 East 29th Street, New York City, NY USA
| |
Collapse
|
29
|
Gillen M, Yang C, Wilson D, Valdez S, Lee C, Kerr B, Shen Z. Evaluation of Pharmacokinetic Interactions Between Lesinurad, a New Selective Urate Reabsorption Inhibitor, and CYP Enzyme Substrates Sildenafil, Amlodipine, Tolbutamide, and Repaglinide. Clin Pharmacol Drug Dev 2017; 6:363-376. [DOI: 10.1002/cpdd.324] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/02/2016] [Indexed: 01/05/2023]
Affiliation(s)
| | - Chun Yang
- Ardea Biosciences, Inc.; San Diego CA USA
| | | | | | | | | | | |
Collapse
|
30
|
Templeton IE, Chen Y, Mao J, Lin J, Yu H, Peters S, Shebley M, Varma MV. Quantitative Prediction of Drug-Drug Interactions Involving Inhibitory Metabolites in Drug Development: How Can Physiologically Based Pharmacokinetic Modeling Help? CPT Pharmacometrics Syst Pharmacol 2016; 5:505-515. [PMID: 27642087 PMCID: PMC5080647 DOI: 10.1002/psp4.12110] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/02/2016] [Accepted: 08/08/2016] [Indexed: 12/26/2022] Open
Abstract
This subteam under the Drug Metabolism Leadership Group (Innovation and Quality Consortium) investigated the quantitative role of circulating inhibitory metabolites in drug-drug interactions using physiologically based pharmacokinetic (PBPK) modeling. Three drugs with major circulating inhibitory metabolites (amiodarone, gemfibrozil, and sertraline) were systematically evaluated in addition to the literature review of recent examples. The application of PBPK modeling in drug interactions by inhibitory parent-metabolite pairs is described and guidance on strategic application is provided.
Collapse
Affiliation(s)
| | - Y Chen
- Genentech, South San Francisco, California, USA
| | - J Mao
- Genentech, South San Francisco, California, USA
| | - J Lin
- Pfizer Inc., Groton, Connecticut, USA
| | - H Yu
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | | | - M Shebley
- AbbVie Inc., North Chicago, Illinois, USA
| | - M V Varma
- Pfizer Inc., Groton, Connecticut, USA.
| |
Collapse
|
31
|
Kim SJ, Yoshikado T, Ieiri I, Maeda K, Kimura M, Irie S, Kusuhara H, Sugiyama Y. Clarification of the Mechanism of Clopidogrel-Mediated Drug-Drug Interaction in a Clinical Cassette Small-dose Study and Its Prediction Based on In Vitro Information. Drug Metab Dispos 2016; 44:1622-32. [PMID: 27457785 DOI: 10.1124/dmd.116.070276] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/22/2016] [Indexed: 11/22/2022] Open
Abstract
Clopidogrel is reported to be associated with cerivastatin-induced rhabdomyolysis, and clopidogrel and its metabolites are capable of inhibiting CYP2C8 and OATP 1B1 in vitro. The objective of the present study was to identify the mechanism of clopidogrel-mediated drug-drug interactions (DDIs) on the pharmacokinetics of OATP1B1 and/or CYP2C8 substrates in vivo. A clinical cassette small-dose study using OATPs, CYP2C8, and OATP1B1/CYP2C8 probe drugs (pitavastatin, pioglitazone, and repaglinide, respectively) with or without the coadministration of either 600 mg rifampicin (an inhibitor for OATPs), 200 mg trimethoprim (an inhibitor for CYP2C8), or 300 mg clopidogrel was performed, and the area under the concentration-time curve (AUC) ratios (AUCRs) for probe substrates were predicted using a static model. Clopidogrel increased the AUC of pioglitazone (2.0-fold) and repaglinide (3.1-fold) but did not significantly change the AUC of pitavastatin (1.1-fold). In addition, the AUC of pioglitazone M4, a CYP2C8-mediated metabolite of pioglitazone, was reduced to 70% of the control by coadministration of clopidogrel. The predicted AUCRs using the mechanism-based inhibition of CYP2C8 by clopidogrel acyl-β-glucuronide were similar to the observed AUCRs, and the predicted AUCR (1.1) of repaglinide using only the inhibition of OATP1B1 did not reach the observed AUCR (3.1). In conclusion, a single 300 mg of clopidogrel mainly inhibits CYP2C8-mediated metabolism by clopidogrel acyl-β-glucuronide, but its effect on the pharmacokinetics of OATP1B1 substrates is negligible. Clopidogrel is expected to have an effect not only on CYP2C8 substrates, but also dual CYP2C8/OATP1B1 substrates as seen in the case of repaglinide.
Collapse
Affiliation(s)
- Soo-Jin Kim
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| | - Takashi Yoshikado
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| | - Ichiro Ieiri
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| | - Kazuya Maeda
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| | - Miyuki Kimura
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| | - Shin Irie
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| | - Hiroyuki Kusuhara
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN, Yokohama, Japan (S. K., T.Y., Y.S.); Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.); Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.M., H.K.); and Sugioka Memorial Hospital, Fukuoka, Japan (M.K., S.I.)
| |
Collapse
|
32
|
Tornio A, Neuvonen PJ, Niemi M, Backman JT. Role of gemfibrozil as an inhibitor of CYP2C8 and membrane transporters. Expert Opin Drug Metab Toxicol 2016; 13:83-95. [PMID: 27548563 DOI: 10.1080/17425255.2016.1227791] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Cytochrome P450 (CYP) 2C8 is a drug metabolizing enzyme of major importance. The lipid-lowering drug gemfibrozil has been identified as a strong inhibitor of CYP2C8 in vivo. This effect is due to mechanism-based inhibition of CYP2C8 by gemfibrozil 1-O-β-glucuronide. In vivo, gemfibrozil is a fairly selective CYP2C8 inhibitor, which lacks significant inhibitory effect on other CYP enzymes. Gemfibrozil can, however, have a smaller but clinically meaningful inhibitory effect on membrane transporters, such as organic anion transporting polypeptide 1B1 and organic anion transporter 3. Areas covered: This review describes the inhibitory effects of gemfibrozil on CYP enzymes and membrane transporters. The clinical drug interactions caused by gemfibrozil and the different mechanisms contributing to the interactions are reviewed in detail. Expert opinion: Gemfibrozil is a useful probe inhibitor of CYP2C8 in vivo, but its effect on membrane transporters has to be taken into account in study design and interpretation. Moreover, gemfibrozil could be used to boost the pharmacokinetics of CYP2C8 substrate drugs. Identification of gemfibrozil 1-O-β-glucuronide as a potent mechanism-based inhibitor of CYP2C8 has led to recognition of glucuronide metabolites as perpetrators of drug-drug interactions. Recently, also acyl glucuronide metabolites of clopidogrel and deleobuvir have been shown to strongly inhibit CYP2C8.
Collapse
Affiliation(s)
- Aleksi Tornio
- a Department of Clinical Pharmacology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Pertti J Neuvonen
- a Department of Clinical Pharmacology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Mikko Niemi
- a Department of Clinical Pharmacology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Janne T Backman
- a Department of Clinical Pharmacology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| |
Collapse
|
33
|
Itkonen MK, Tornio A, Neuvonen M, Neuvonen PJ, Niemi M, Backman JT. Clopidogrel Markedly Increases Plasma Concentrations of CYP2C8 Substrate Pioglitazone. Drug Metab Dispos 2016; 44:1364-71. [PMID: 27260150 DOI: 10.1124/dmd.116.070375] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/02/2016] [Indexed: 01/20/2023] Open
Abstract
The glucose-lowering drug pioglitazone undergoes hepatic CYP2C8-mediated biotransformation to its main metabolites. The antiplatelet drug clopidogrel is metabolized to clopidogrel acyl-β-d-glucuronide, which was recently found to be a strong time-dependent inhibitor of CYP2C8 in humans. Therefore, we studied the effect of clopidogrel on the pharmacokinetics of pioglitazone. In a randomized crossover study, 10 healthy volunteers ingested either 300 mg of clopidogrel on day 1, and 75 mg on days 2 and 3, or placebo. Pioglitazone 15 mg was administered 1 hour after placebo and clopidogrel on day 1. Plasma concentrations of pioglitazone, clopidogrel, and their main metabolites were measured up to 72 hours. Clopidogrel increased the area under the plasma concentration-time curve (AUC0-∞) of pioglitazone 2.1-fold [P < 0.001, 90% confidence interval (CI) 1.8-2.6] and prolonged its half-life from 6.7 to 11 hours (P = 0.002). The peak concentration of pioglitazone was unaffected but the concentration at 24 hours was increased 4.5-fold (range 1.6-9.8; P < 0.001, 90% CI 3.17-6.45) by clopidogrel. The M-IV-to-pioglitazone AUC0-∞ ratio was 49% (P < 0.001, 90% CI 0.40-0.59) of that during the control phase, indicating that clopidogrel inhibited the CYP2C8-mediated biotransformation of pioglitazone. Clopidogrel increases the exposure to pioglitazone by inhibiting its CYP2C8-mediated biotransformation. In consequence, use of clopidogrel may increase the risk of fluid retention and other concentration-related adverse effects of pioglitazone.
Collapse
Affiliation(s)
- Matti K Itkonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
34
|
CYP2C8-mediated interaction between repaglinide and steviol acyl glucuronide: In vitro investigations using rat and human matrices and in vivo pharmacokinetic evaluation in rats. Food Chem Toxicol 2016; 94:138-47. [DOI: 10.1016/j.fct.2016.05.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/28/2016] [Accepted: 05/31/2016] [Indexed: 01/01/2023]
|
35
|
Foti RS, Diaz P, Douguet D. Comparison of the ligand binding site of CYP2C8 with CYP26A1 and CYP26B1: a structural basis for the identification of new inhibitors of the retinoic acid hydroxylases. J Enzyme Inhib Med Chem 2016; 31:148-161. [PMID: 27424662 DOI: 10.1080/14756366.2016.1193734] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The CYP26s are responsible for metabolizing retinoic acid and play an important role in maintaining homeostatic levels of retinoic acid. Given the ability of CYP2C8 to metabolize retinoic acid, we evaluated the potential for CYP2C8 inhibitors to also inhibit CYP26. In vitro assays were used to evaluate the inhibition potencies of CYP2C8 inhibitors against CYP26A1 and CYP26B1. Using tazarotenic acid as a substrate for CYP26, IC50 values for 17 inhibitors of CYP2C8 were determined for CYP26A1 and CYP26B1, ranging from ∼20 nM to 100 μM, with a positive correlation observed between IC50s for CYP2C8 and CYP26A1. An evaluation of IC50's versus in vivo Cmax values suggests that inhibitors such as clotrimazole or fluconazole may interact with CYP26 at clinically relevant concentrations and may alter levels of retinoic acid. These findings provide insight into drug interactions resulting in elevated retinoic acid concentrations and expand upon the pharmacophore of CYP26 inhibition.
Collapse
Affiliation(s)
- Robert S Foti
- a Amgen Pharmacokinetics and Drug Metabolism , Cambridge , MA , USA
| | - Philippe Diaz
- b Department of Biomedical and Pharmaceutical Sciences , Core Laboratory for Neuromolecular Production, University of Montana , Missoula , MT , USA.,c Dermaxon , Missoula , MT , USA , and
| | - Dominique Douguet
- d CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Nice Sophia Antipolis , Valbonne , France
| |
Collapse
|
36
|
Backman JT, Filppula AM, Niemi M, Neuvonen PJ. Role of Cytochrome P450 2C8 in Drug Metabolism and Interactions. Pharmacol Rev 2016; 68:168-241. [PMID: 26721703 DOI: 10.1124/pr.115.011411] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
During the last 10-15 years, cytochrome P450 (CYP) 2C8 has emerged as an important drug-metabolizing enzyme. CYP2C8 is highly expressed in human liver and is known to metabolize more than 100 drugs. CYP2C8 substrate drugs include amodiaquine, cerivastatin, dasabuvir, enzalutamide, imatinib, loperamide, montelukast, paclitaxel, pioglitazone, repaglinide, and rosiglitazone, and the number is increasing. Similarly, many drugs have been identified as CYP2C8 inhibitors or inducers. In vivo, already a small dose of gemfibrozil, i.e., 10% of its therapeutic dose, is a strong, irreversible inhibitor of CYP2C8. Interestingly, recent findings indicate that the acyl-β-glucuronides of gemfibrozil and clopidogrel cause metabolism-dependent inactivation of CYP2C8, leading to a strong potential for drug interactions. Also several other glucuronide metabolites interact with CYP2C8 as substrates or inhibitors, suggesting that an interplay between CYP2C8 and glucuronides is common. Lack of fully selective and safe probe substrates, inhibitors, and inducers challenges execution and interpretation of drug-drug interaction studies in humans. Apart from drug-drug interactions, some CYP2C8 genetic variants are associated with altered CYP2C8 activity and exhibit significant interethnic frequency differences. Herein, we review the current knowledge on substrates, inhibitors, inducers, and pharmacogenetics of CYP2C8, as well as its role in clinically relevant drug interactions. In addition, implications for selection of CYP2C8 marker and perpetrator drugs to investigate CYP2C8-mediated drug metabolism and interactions in preclinical and clinical studies are discussed.
Collapse
Affiliation(s)
- Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| | - Anne M Filppula
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| |
Collapse
|
37
|
Kimoto E, Li R, Scialis RJ, Lai Y, Varma MVS. Hepatic Disposition of Gemfibrozil and Its Major Metabolite Gemfibrozil 1-O-β-Glucuronide. Mol Pharm 2015; 12:3943-52. [DOI: 10.1021/acs.molpharmaceut.5b00411] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Emi Kimoto
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Rui Li
- Systems
Modeling and Simulation, Department of Pharmacokinetics, Dynamics,
and Metabolism, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, United States
| | - Renato J. Scialis
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Yurong Lai
- Departments
of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, , Princeton, New Jersey 08540, United States
| | - Manthena V. S. Varma
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| |
Collapse
|
38
|
Zhao RY, He XW, Shan YM, Zhu LL, Zhou Q. A stewardship intervention program for safe medication management and use of antidiabetic drugs. Clin Interv Aging 2015; 10:1201-12. [PMID: 26229454 PMCID: PMC4516029 DOI: 10.2147/cia.s87456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Diabetes patients are complex due to considerations of polypharmacy, multimorbidities, medication adherence, dietary habits, health literacy, socioeconomic status, and cultural factors. Meanwhile, insulin and oral hypoglycemic agents are high-alert medications. Therefore it is necessary to require a multidisciplinary team’s integrated endeavors to enhance safe medication management and use of antidiabetic drugs. Methods A 5-year stewardship intervention program, including organizational measures and quality improvement activities in storage, prescription, dispensing, administration, and monitoring, was performed in the Second Affiliated Hospital of Zhejiang University, People’s Republic of China, a 3,200-bed hospital with 3.5 million outpatient visits annually. Results The Second Affiliated Hospital of Zhejiang University has obtained a 100% implementation rate of standard storage of antidiabetic drugs in the Pharmacy and wards since August 2012. A zero occurrence of dispensing errors related to highly “look-alike” and “sound-alike” NovoMix 30® (biphasic insulin aspart) and NovoRapid® (insulin aspart) has been achieved since October 2011. Insulin injection accuracy among ward nurses significantly increased from 82% (first quarter 2011) to 96% (fourth quarter 2011) (P<0.05). The number of medication administration errors related to insulin continuously decreased from 20 (2011) to six (2014). The occurrence rate of hypoglycemia in non–endocrinology ward diabetes inpatients during 2011–2013 was significantly less than that in 2010 (5.03%–5.53% versus 8.27%) (P<0.01). Percentage of correct management of hypoglycemia by nurses increased from 41.5% (April 2014) to 67.2% (August 2014) (P<0.01). The percentage of outpatient diabetes patients receiving standard insulin injection education increased from 80% (April 2012) to 95.2% (October 2012) (P<0.05). Insulin injection techniques among diabetes outpatients who started to receive insulin were better than indicated in data from two questionnaire surveys in the literature, including the percentage checking injection sites prior to injection (85.6%), priming before injection (98.1%), rotation of injecting sites (98.1%), remixing before use (94.5%), keeping the pen needle under the skin for >10 seconds (99.4%), and using the pen needle only once (88.7%). On-site inspection indicated of great improvement in the percentage of drug-related problems in the antidiabetes regimen between the first and second quarter of 2014 (1.08% versus 0.28%) (P<0.05). Conclusion Quality improvements in safe medication management and use of antidiabetic drugs can be achieved by multidisciplinary collaboration among pharmacists, nurses, physicians, and information engineers.
Collapse
Affiliation(s)
- Rui-yi Zhao
- Clinical Nurse Specialist Section, Division of Nursing, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xiao-wen He
- Clinical Nurse Specialist Section, Division of Nursing, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yan-min Shan
- Clinical Nurse Specialist Section, Division of Nursing, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Ling-ling Zhu
- Geriatric VIP Care Ward, Division of Nursing, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Quan Zhou
- Department of Pharmacy, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
39
|
Varma MVS, Lin J, Bi YA, Kimoto E, Rodrigues AD. Quantitative Rationalization of Gemfibrozil Drug Interactions: Consideration of Transporters-Enzyme Interplay and the Role of Circulating Metabolite Gemfibrozil 1-O-β-Glucuronide. Drug Metab Dispos 2015; 43:1108-18. [PMID: 25941268 DOI: 10.1124/dmd.115.064303] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/04/2015] [Indexed: 01/06/2023] Open
Abstract
Gemfibrozil has been suggested as a sensitive cytochrome P450 2C8 (CYP2C8) inhibitor for clinical investigation by the U.S. Food and Drug Administration and the European Medicines Agency. However, gemfibrozil drug-drug interactions (DDIs) are complex; its major circulating metabolite, gemfibrozil 1-O-β-glucuronide (Gem-Glu), exhibits time-dependent inhibition of CYP2C8, and both parent and metabolite also behave as moderate inhibitors of organic anion transporting polypeptide 1B1 (OATP1B1) in vitro. Additionally, parent and metabolite also inhibit renal transport mediated by OAT3. Here, in vitro inhibition data for gemfibrozil and Gem-Glu were used to assess their impact on the pharmacokinetics of several victim drugs (including rosiglitazone, pioglitazone, cerivastatin, and repaglinide) by employing both static mechanistic and dynamic physiologically based pharmacokinetic (PBPK) models. Of the 48 cases evaluated using the static models, about 75% and 98% of the DDIs were predicted within 1.5- and 2-fold of the observed values, respectively, when incorporating the interaction potential of both gemfibrozil and its 1-O-β-glucuronide. Moreover, the PBPK model was able to recover the plasma profiles of rosiglitazone, pioglitazone, cerivastatin, and repaglinide under control and gemfibrozil treatment conditions. Analyses suggest that Gem-Glu is the major contributor to the DDIs, and its exposure needed to bring about complete inactivation of CYP2C8 is only a fraction of that achieved in the clinic after a therapeutic gemfibrozil dose. Overall, the complex interactions of gemfibrozil can be quantitatively rationalized, and the learnings from this analysis can be applied in support of future predictions of gemfibrozil DDIs.
Collapse
Affiliation(s)
- Manthena V S Varma
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Jian Lin
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Yi-an Bi
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Emi Kimoto
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - A David Rodrigues
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
40
|
Varma MV, Pang KS, Isoherranen N, Zhao P. Dealing with the complex drug-drug interactions: Towards mechanistic models. Biopharm Drug Dispos 2015; 36:71-92. [DOI: 10.1002/bdd.1934] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/11/2014] [Accepted: 12/14/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Manthena V. Varma
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Inc; Groton Connecticut USA
| | - K. Sandy Pang
- Leslie Dan Faculty of Pharmacy; University of Toronto; M5S 3M2 Canada
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy; University of Washington; Seattle WA USA
| | - Ping Zhao
- Division of Pharmacometrics, Office of Clinical Pharmacology/Office of Translational Sciences; Center for Drug Evaluation and Research, US Food and Drug Administration; Silver Spring MD USA
| |
Collapse
|
41
|
Glucuronidation converts clopidogrel to a strong time-dependent inhibitor of CYP2C8: a phase II metabolite as a perpetrator of drug-drug interactions. Clin Pharmacol Ther 2014; 96:498-507. [PMID: 24971633 DOI: 10.1038/clpt.2014.141] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/18/2014] [Indexed: 12/22/2022]
Abstract
Cerivastatin and repaglinide are substrates of cytochrome P450 (CYP)2C8, CYP3A4, and organic anion-transporting polypeptide (OATP)1B1. A recent study revealed an increased risk of rhabdomyolysis in patients using cerivastatin with clopidogrel, warranting further studies on clopidogrel interactions. In healthy volunteers, repaglinide area under the concentration-time curve (AUC(0-∞)) was increased 5.1-fold by a 300-mg loading dose of clopidogrel and 3.9-fold by continued administration of 75 mg clopidogrel daily. In vitro, we identified clopidogrel acyl-β-D-glucuronide as a potent time-dependent inhibitor of CYP2C8. A physiologically based pharmacokinetic model indicated that inactivation of CYP2C8 by clopidogrel acyl-β-D-glucuronide leads to uninterrupted 60-85% inhibition of CYP2C8 during daily clopidogrel treatment. Computational modeling resulted in docking of clopidogrel acyl-β-D-glucuronide at the CYP2C8 active site with its thiophene moiety close to heme. The results indicate that clopidogrel is a strong CYP2C8 inhibitor via its acyl-β-D-glucuronide and imply that glucuronide metabolites should be considered potential inhibitors of CYP enzymes.
Collapse
|
42
|
PharmGKB summary: very important pharmacogene information for cytochrome P450, family 2, subfamily C, polypeptide 8. Pharmacogenet Genomics 2014; 23:721-8. [PMID: 23962911 DOI: 10.1097/fpc.0b013e3283653b27] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Gertz M, Tsamandouras N, Säll C, Houston JB, Galetin A. Reduced physiologically-based pharmacokinetic model of repaglinide: impact of OATP1B1 and CYP2C8 genotype and source of in vitro data on the prediction of drug-drug interaction risk. Pharm Res 2014; 31:2367-82. [PMID: 24623479 DOI: 10.1007/s11095-014-1333-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 02/08/2014] [Indexed: 12/15/2022]
Abstract
PURPOSE To investigate the effect of OATP1B1 genotype as a covariate on repaglinide pharmacokinetics and drug-drug interaction (DDIs) risk using a reduced physiologically-based pharmacokinetic (PBPK) model. METHODS Twenty nine mean plasma concentration-time profiles for SLCO1B1 c.521T>C were used to estimate hepatic uptake clearance (CLuptake) in different genotype groups applying a population approach in NONMEM v.7.2. RESULTS Estimated repaglinide CLuptake corresponded to 217 and 113 μL/min/10(6) cells for SLCO1B1 c.521TT/TC and CC, respectively. A significant effect of OATP1B1 genotype was seen on CLuptake (48% reduction for CC relative to wild type). Sensitivity analysis highlighted the impact of CLmet and CLdiff uncertainty on the CLuptake optimization using plasma data. Propagation of this uncertainty had a marginal effect on the prediction of repaglinide OATP1B1-mediated DDI with cyclosporine; however, sensitivity of the predicted magnitude of repaglinide metabolic DDI was high. In addition, the reduced PBPK model was used to assess the effect of both CYP2C8*3 and SLCO1B1 c.521T>C on repaglinide exposure by simulations; power calculations were performed to guide prospective DDI and pharmacogenetic studies. CONCLUSIONS The application of reduced PBPK model for parameter optimization and limitations of this process associated with the use of plasma rather than tissue profiles are illustrated.
Collapse
Affiliation(s)
- Michael Gertz
- Centre for Applied Pharmacokinetic Research Manchester Pharmacy School, The University of Manchester, Oxford Road, M13 9PT, Manchester, UK
| | | | | | | | | |
Collapse
|
44
|
Abstract
This chapter describes the types of irreversible inhibition of drug-metabolizing enzymes and the methods commonly employed to quantify the irreversible inhibition and subsequently predict the extent and time course of clinically important drug-drug interactions.
Collapse
Affiliation(s)
- Michael Mohutsky
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | |
Collapse
|
45
|
Stresser DM, Mao J, Kenny JR, Jones BC, Grime K. Exploring concepts ofin vitrotime-dependent CYP inhibition assays. Expert Opin Drug Metab Toxicol 2013; 10:157-74. [DOI: 10.1517/17425255.2014.856882] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
46
|
Aquilante CL, Kosmiski LA, Bourne DWA, Bushman LR, Daily EB, Hammond KP, Hopley CW, Kadam RS, Kanack AT, Kompella UB, Le M, Predhomme JA, Rower JE, Sidhom MS. Impact of the CYP2C8 *3 polymorphism on the drug-drug interaction between gemfibrozil and pioglitazone. Br J Clin Pharmacol 2013; 75:217-26. [PMID: 22625877 DOI: 10.1111/j.1365-2125.2012.04343.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
AIM The objective of this study was to determine the extent to which the CYP2C8*3 allele influences pharmacokinetic variability in the drug-drug interaction between gemfibrozil (CYP2C8 inhibitor) and pioglitazone (CYP2C8 substrate). METHODS In this randomized, two phase crossover study, 30 healthy Caucasian subjects were enrolled based on CYP2C8*3 genotype (n = 15, CYP2C8*1/*1; n = 15, CYP2C8*3 carriers). Subjects received a single 15 mg dose of pioglitazone or gemfibrozil 600 mg every 12 h for 4 days with a single 15 mg dose of pioglitazone administered on the morning of day 3. A 48 h pharmacokinetic study followed each pioglitazone dose and the study phases were separated by a 14 day washout period. RESULTS Gemfibrozil significantly increased mean pioglitazone AUC(0,∞) by 4.3-fold (P < 0.001) and there was interindividual variability in the magnitude of this interaction (range, 1.8- to 12.1-fold). When pioglitazone was administered alone, the mean AUC(0,∞) was 29.7% lower (P = 0.01) in CYP2C8*3 carriers compared with CYP2C8*1 homozygotes. The relative change in pioglitazone plasma exposure following gemfibrozil administration was significantly influenced by CYP2C8 genotype. Specifically, CYP2C8*3 carriers had a 5.2-fold mean increase in pioglitazone AUC(0,∞) compared with a 3.3-fold mean increase in CYP2C8*1 homozygotes (P = 0.02). CONCLUSION CYP2C8*3 is associated with decreased pioglitazone plasma exposure in vivo and significantly influences the pharmacokinetic magnitude of the gemfibrozil-pioglitazone drug-drug interaction. Additional studies are needed to evaluate the impact of CYP2C8 genetics on the pharmacokinetics of other CYP2C8-mediated drug-drug interactions.
Collapse
Affiliation(s)
- Christina L Aquilante
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Filppula AM, Tornio A, Niemi M, Neuvonen PJ, Backman JT. Gemfibrozil Impairs Imatinib Absorption and Inhibits the CYP2C8-Mediated Formation of Its Main Metabolite. Clin Pharmacol Ther 2013; 94:383-93. [DOI: 10.1038/clpt.2013.92] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/01/2013] [Indexed: 12/14/2022]
|
48
|
Varma MVS, Lin J, Bi YA, Rotter CJ, Fahmi OA, Lam JL, El-Kattan AF, Goosen TC, Lai Y. Quantitative prediction of repaglinide-rifampicin complex drug interactions using dynamic and static mechanistic models: delineating differential CYP3A4 induction and OATP1B1 inhibition potential of rifampicin. Drug Metab Dispos 2013; 41:966-74. [PMID: 23393219 DOI: 10.1124/dmd.112.050583] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Repaglinide is mainly metabolized by cytochrome P450 enzymes CYP2C8 and CYP3A4, and it is also a substrate to a hepatic uptake transporter, organic anion transporting polypeptide (OATP)1B1. The purpose of this study is to predict the dosing time-dependent pharmacokinetic interactions of repaglinide with rifampicin, using mechanistic models. In vitro hepatic transport of repaglinide, characterized using sandwich-cultured human hepatocytes, and intrinsic metabolic parameters were used to build a dynamic whole-body physiologically-based pharmacokinetic (PBPK) model. The PBPK model adequately described repaglinide plasma concentration-time profiles and successfully predicted area under the plasma concentration-time curve ratios of repaglinide (within ± 25% error), dosed (staggered 0-24 hours) after rifampicin treatment when primarily considering induction of CYP3A4 and reversible inhibition of OATP1B1 by rifampicin. Further, a static mechanistic "extended net-effect" model incorporating transport and metabolic disposition parameters of repaglinide and interaction potency of rifampicin was devised. Predictions based on the static model are similar to those observed in the clinic (average error ∼19%) and to those based on the PBPK model. Both the models suggested that the combined effect of increased gut extraction and decreased hepatic uptake caused minimal repaglinide systemic exposure change when repaglinide is dosed simultaneously or 1 hour after the rifampicin dose. On the other hand, isolated induction effect as a result of temporal separation of the two drugs translated to an approximate 5-fold reduction in repaglinide systemic exposure. In conclusion, both dynamic and static mechanistic models are instrumental in delineating the quantitative contribution of transport and metabolism in the dosing time-dependent repaglinide-rifampicin interactions.
Collapse
Affiliation(s)
- Manthena V S Varma
- Department of Pharmacokinetics, Dynamics, and Metabolism, MS 8220-2451, Pfizer World Wide Research and Development, Pfizer Inc., Groton, CT 06340, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Characterization of the circulating metabolites for a new chemical entity in humans is essential for safety assessment, an understanding of their contributions to pharmacologic activities, and their potential involvement in drug-drug interactions. This review examines the abundance of metabolites relative to the total parent drug [metabolite-to-parent (M/P) ratio] from 125 drugs in relation to their structural and physicochemical characteristics, lipoidal permeability, protein binding, and fractional formation from parent (fm). Our analysis suggests that fm is the major determinant of total drug M/P ratio for amine, alcohol, N- and S-oxide, and carboxylic acid metabolites. Passage from the hepatocyte to systemic circulation does not appear to be limiting owing to the vast majority of metabolites formed being relatively lipid permeable. In some cases, active transport plays an important role in this process (e.g., carboxylic acid metabolites). Differences in total parent drug clearance and metabolite clearance are attenuated by the reduction in lipophilicity introduced by the metabolic step and resultant compensatory changes in unbound clearance and protein binding. A small subclass of these drugs (e.g., terfenadine) is unintentional prodrugs with very high parent drug clearance, resulting in very high M/P ratios. In contrast, arenol metabolites show a more complex relationship with fm due largely to the new metabolic routes (conjugation) available to the metabolite compared with the parent drug molecule. For these metabolites, a more thorough understanding of the elimination clearance of the metabolite is critical to discern the likelihood of whether the phenol will constitute a major circulating metabolite.
Collapse
Affiliation(s)
- Cho-Ming Loi
- Pfizer, Inc., 10646 Science Center Drive, San Diego, CA 92121, USA.
| | | | | |
Collapse
|
50
|
Holstein A, Beil W, Kovacs P. CYP2C metabolism of oral antidiabetic drugs--impact on pharmacokinetics, drug interactions and pharmacogenetic aspects. Expert Opin Drug Metab Toxicol 2013; 8:1549-63. [PMID: 23153186 DOI: 10.1517/17425255.2012.722619] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The cytochrome P4502C enzymes account for the metabolism of approximately 20% of therapeutic drugs including certain oral antidiabetic drugs (OADs). AREAS COVERED This review focuses on the effect of CYP2C enzymes on metabolism of sulphonylureas (SUs), meglitinides, and thiazolidinediones (TZDs) discussing their impact on pharmacokinetics, drug interactions and toxicological profiles. Pharmacogenetic aspects reflecting individual gene variants and variable drug effects are also considered. EXPERT OPINION Genetic polymorphisms of CYP2C9 enzymes (*2/*2, *2/*3, *3/*3) influence the glycaemic response to SUs and impair their substrate metabolism. Restricted data from small-sized studies with heterogenous definitions of hypoglycaemia revealed no clear association between CYP2C9 genotypes and the risk of hypoglycaemia. Functional polymorphisms of CYP2C8- and CYP2C9 drug metabolizing genes affect markedly pharmacokinetics of meglitinides. Compared to wild-type carriers, patients treated with TZDs and carrying the common CYP2C8*3 and *4 variants showed a reduced glycaemic control. The strong CYP2C8 and OATP1B1 inhibitor gemfibrozil increases substantially the plasma concentrations of repaglinide and TZDs. Numerous metabolic drug interactions exist between SUs and commonly prescribed drugs, especially anti-infectives. The complex pharmacokinetic and pharmacogenetic properties and the unfavourable short and long term risk profile of glibenclamide and glimepiride raise the question whether their use can be justified any longer.
Collapse
Affiliation(s)
- Andreas Holstein
- Lippe-Detmold Hospital, First Department of Medicine, Röntgenstr. 18, Detmold, 32756, Germany.
| | | | | |
Collapse
|