51
|
Lutz JD, Kirby BJ, Wang L, Song Q, Ling J, Massetto B, Worth A, Kearney BP, Mathias A. Cytochrome P450 3A Induction Predicts P-glycoprotein Induction; Part 1: Establishing Induction Relationships Using Ascending Dose Rifampin. Clin Pharmacol Ther 2018; 104:1182-1190. [PMID: 29569723 PMCID: PMC6282691 DOI: 10.1002/cpt.1073] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/26/2018] [Accepted: 03/03/2018] [Indexed: 11/06/2022]
Abstract
Drug transporter and cytochrome P450 expression is regulated by shared nuclear receptors and, hence, an inducer should induce both, although the magnitude may differ. The objective of this study was to establish relative induction relationships between CYP3A and drug transporters (P-glycoprotein (P-gp), organic anion transporting polypeptide (OATP), and breast cancer resistance protein (BCRP)) or other P450s (CYP2C9 and CYP1A2) using ascending doses of the prototypical pregnane xenobiotic receptor (PXR) agonist, rifampin, to elicit weak, moderate, and strong PXR agonism. Healthy subjects received dabigatran etexilate, pravastatin, rosuvastatin, and a midazolam/tolbutamide/caffeine cocktail before and after rifampin 2, 10, 75, or 600 mg q.d. Unlike CYP3A, only moderate induction of P-gp, OATP, and CYP2C9 was observed and dose-dependent induction of P-gp, OATP, and CYP2C9 was always one drug-drug interaction category lower than observed for CYP3A, even when correcting for probe drug sensitivity. Data from this study establish proof-of-concept that P450 induction data can be leveraged to inform on the effect on transporters.
Collapse
Affiliation(s)
- Justin D Lutz
- Department of Clinical Pharmacology, Gilead Sciences, Inc., Foster City, California, USA
| | - Brian J Kirby
- Department of Clinical Pharmacology, Gilead Sciences, Inc., Foster City, California, USA
| | - Lu Wang
- Department of Biometrics, Gilead Sciences, Inc., Foster City, California, USA
| | - Qinghua Song
- Department of Biometrics, Gilead Sciences, Inc., Foster City, California, USA
| | - John Ling
- Department of Clinical Pharmacology, Gilead Sciences, Inc., Foster City, California, USA
| | - Benedetta Massetto
- Department of Clinical Operations, Gilead Sciences, Inc., Foster City, California, USA
| | - Angela Worth
- Department of Clinical Research, Gilead Sciences, Inc., Foster City, California, USA
| | - Brian P Kearney
- Department of Clinical Pharmacology, Gilead Sciences, Inc., Foster City, California, USA
| | - Anita Mathias
- Department of Clinical Pharmacology, Gilead Sciences, Inc., Foster City, California, USA
| |
Collapse
|
52
|
Moltó J, Rajoli R, Back D, Valle M, Miranda C, Owen A, Clotet B, Siccardi M. Use of a physiologically based pharmacokinetic model to simulate drug-drug interactions between antineoplastic and antiretroviral drugs. J Antimicrob Chemother 2017; 72:805-811. [PMID: 27999009 DOI: 10.1093/jac/dkw485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 10/10/2016] [Indexed: 11/13/2022] Open
Abstract
Background Co-administration of antineoplastics with ART is challenging due to potential drug-drug interactions (DDIs). However, trials specifically assessing such DDIs are lacking. Our objective was to simulate DDIs between the antineoplastics erlotinib and gefitinib with key antiretroviral drugs and to predict dose adjustments using a physiologically based pharmacokinetic (PBPK) model. Methods In vitro data describing chemical properties and pharmacokinetic processes of each drug and their effect on cytochrome P450 isoforms were obtained from the literature. Plasma drug-concentration profiles were simulated in a virtual population of 50 individuals receiving erlotinib or gefitinib alone or with darunavir/ritonavir, efavirenz or etravirine. Simulated pharmacokinetic parameters and the magnitude of DDIs with probe drugs (midazolam, maraviroc) were compared with literature values. Erlotinib and gefitinib pharmacokinetics with and without antiretrovirals were compared and dose-adjustment strategies were evaluated. Results Simulated parameters of each drug and the magnitude of DDIs with probe drugs were in agreement with reference values. Darunavir/ritonavir increased erlotinib and gefitinib exposure, while efavirenz and etravirine decreased erlotinib and gefitinib concentrations. Based on our predictions, dose-adjustment strategies may consist of once-daily dosing erlotinib at 25 mg and gefitinib at 125 mg with darunavir/ritonavir; or erlotinib at 200 mg and gefitinib at 375 mg with etravirine. The interaction with efavirenz was not overcome even after doubling erlotinib or gefitinib doses. Conclusions PBPK models predicted the in vivo pharmacokinetics of erlotinib, gefitinib and the antiretrovirals darunavir/ritonavir, efavirenz and etravirine, and the DDIs between them. The simulated dose-adjustments may represent valuable strategies to optimize antineoplastic therapy in HIV-infected patients.
Collapse
Affiliation(s)
- José Moltó
- Lluita contra la Sida Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Universitat Autónoma de Barcelona (UAB), Barcelona, Spain
| | - Rajith Rajoli
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - David Back
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Marta Valle
- Universitat Autónoma de Barcelona (UAB), Barcelona, Spain.,PKPD Modeling and Simulation, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau - Institut d'Investigacio Biomedica Sant Pau, Barcelona, Spain
| | - Cristina Miranda
- Lluita contra la Sida Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Andrew Owen
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Bonaventura Clotet
- Lluita contra la Sida Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Universitat Autónoma de Barcelona (UAB), Barcelona, Spain.,Universitat de Vic (UVic), Vic, Spain.,IrsiCaixa Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Marco Siccardi
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| |
Collapse
|
53
|
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
|
54
|
Hossain MA, Tran T, Chen T, Mikus G, Greenblatt DJ. Inhibition of human cytochromes P450 in vitro by ritonavir and cobicistat. J Pharm Pharmacol 2017; 69:1786-1793. [PMID: 28960344 DOI: 10.1111/jphp.12820] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/26/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Ritonavir and cobicistat are strong inhibitors of human cytochrome P450-3A (CYP3A) isoforms, and are used clinically as pharmacokinetic boosting agents for other antiretroviral drugs. Data reported by the manufacturer suggest that cobicistat is a more selective inhibitor of CYP3A than ritonavir. However, this claim has not been validated in clinical studies. This study evaluated the in-vitro inhibitory potency of ritonavir and cobicistat vs a series of human CYP isoforms. METHOD The model system utilized human liver microsomes and isoform-selective index substrates. KEY FINDINGS Ritonavir and cobicistat both were strong inhibitors of CYP3A4, with IC50 values of 0.014 and 0.032 μm, respectively. A component of inhibition was time-dependent (mechanism-based). Neither drug meaningfully inhibited CYP1A2 (IC50 > 150 μm). CYP2B6, CYP2C9, CYP2C19 and CYP2D6 were inhibited by both drugs, but with IC50 values exceeding 6 μm. CONCLUSIONS Consistent with previous reports, both ritonavir and cobicistat were highly potent inhibitors of CYP3A. Both drugs were weaker inhibitors of other human CYPs, with IC50 values at least two orders of magnitude higher. There was no evidence of a meaningful difference in selectivity between the two drugs.
Collapse
Affiliation(s)
- Md Amin Hossain
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Timothy Tran
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Tianmeng Chen
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - David J Greenblatt
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.,Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
55
|
Midde NM, Gong Y, Cory TJ, Li J, Meibohm B, Li W, Kumar S. Influence of Ethanol on Darunavir Hepatic Clearance and Intracellular PK/PD in HIV-Infected Monocytes, and CYP3A4-Darunavir Interactions Using Inhibition and in Silico Binding Studies. Pharm Res 2017; 34:1925-1933. [PMID: 28616684 DOI: 10.1007/s11095-017-2203-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/05/2017] [Indexed: 02/08/2023]
Abstract
PURPOSE Although the prevalence of alcohol consumption is higher in HIV+ people than general public, limited information is available on how alcohol affects the metabolism and bioavailability of darunavir (DRV). METHODS DRV was quantified by using LC-MS/MS method. All in vitro experiments were performed using human liver microsomes and HIV-infected monocytic cells. CYP3A4 and DRV/Ritonavir (RTV) docking was performed using GOLD suite 5.8. RESULTS Ethanol (20 mM) significantly decreased apparent half-life and increased degradation rate constant of RTV-boosted DRV but not for DRV alone. Similarly, ethanol exposure increased hepatic intrinsic clearance for RTV-boosted DRV with no significant influence on DRV alone. Ethanol showed a limited influence on intracellular total DRV exposure in the presence of RTV without altering maximum concentration (Cmax) values in HIV-infected monocytic cells. Ethanol alone elevated HIV replication but this effect was nullified with the addition of DRV or DRV + RTV. Additionally, inhibitory potency of DRV was significantly reduced in the presence of ethanol. Our docking results projected that ethanol increases the average distance between DRV and CYP3A4 heme, and alter the orientation of DRV-CYP3A4 binding. CONCLUSIONS Collectively these findings suggest that DRV metabolism is primarily influenced by ethanol in the liver, but has minor effect in HIV-residing monocytes.
Collapse
Affiliation(s)
- Narasimha M Midde
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Ave, Rm 456, Memphis, TN, 38163, USA.
| | - Yuqing Gong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Ave, Rm 456, Memphis, TN, 38163, USA
| | - Theodore J Cory
- Department of Clinical Pharmacy, College of Pharmacy, University of Tennessee Health Science Center,, Memphis, TN, 38163, USA
| | - Junhao Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology,, Shanghai, 200237, China
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Ave, Rm 456, Memphis, TN, 38163, USA
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology,, Shanghai, 200237, China
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Ave, Rm 456, Memphis, TN, 38163, USA.
| |
Collapse
|
56
|
Tod M, Bourguignon L, Bleyzac N, Goutelle S. A Model for Predicting the Interindividual Variability of Drug-Drug Interactions. AAPS JOURNAL 2016; 19:497-509. [PMID: 27924615 DOI: 10.1208/s12248-016-0021-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/28/2016] [Indexed: 11/30/2022]
Abstract
Pharmacokinetic drug-drug interactions are frequently characterized and quantified by an AUC ratio (Rauc). The typical value of the AUC ratio in case of cytochrome-mediated interactions may be predicted by several approaches, based on in vitro or in vivo data. Prediction of the interindividual variability of Rauc would help to anticipate more completely the consequences of a drug-drug interaction. We propose and evaluate a simple approach for predicting the standard deviation (sd) of Ln(Rauc), a metric close to the interindividual coefficient of variation of Rauc. First, a model was derived to link sd(Ln Rauc) with the substrate fraction metabolized by each cytochrome and the potency of the interactors, in case of induction or inhibition. Second, the parameters involved in these equations were estimated by a Bayesian hierarchical model, using the data from 56 interaction studies retrieved from the literature. Third, the model was evaluated by several metrics based on the fold prediction error (PE) of sd(Ln Rauc). The median PE was 0.998 (the ideal value is 1) and the interquartile range was 0.96-1.03. The PE was in the acceptable interval (0.5 to 2) in 52 cases out of 56. Fourth, a surface plot of sd(Ln Rauc) as a function of the characteristics of the substrate and the interactor has been built. The minimal value of sd(Ln Rauc) was about 0.08 (obtained for Rauc = 1) while the maximal value, 0.7, was obtained for interactions involving highly metabolized substrates with strong interactors.
Collapse
Affiliation(s)
- M Tod
- Pharmacie, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France. .,EMR3738, Faculté de médecine Lyon-sud, Université Lyon 1, Lyon, France. .,Faculté de pharmacie, Université Lyon 1, Lyon, France.
| | - L Bourguignon
- Pharmacie, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France.,Faculté de pharmacie, Université Lyon 1, Lyon, France.,UMR CNRS 5558, Laboratoire de Biométrie et Biologie Evolutive, Université Lyon 1, Lyon, France
| | - N Bleyzac
- EMR3738, Faculté de médecine Lyon-sud, Université Lyon 1, Lyon, France.,Pharmacie, Institut d'Hématologie et d'Oncologie Pédiatrique, Hospices Civils de Lyon, Lyon, France
| | - S Goutelle
- Pharmacie, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France.,Faculté de pharmacie, Université Lyon 1, Lyon, France.,UMR CNRS 5558, Laboratoire de Biométrie et Biologie Evolutive, Université Lyon 1, Lyon, France
| |
Collapse
|
57
|
Lu YY, Cheng HX, Wang X, Wang XW, Liu JY, Li P, Lou YQ, Li J, Lu C, Zhang GL. Identification of cytochrome P450s involved in the metabolism of 6-benzyl-1-benzyloxymethyl-5-iodouracil (W-1) using human recombinant enzymes and rat liver microsomes in vitro. Xenobiotica 2016; 47:667-672. [PMID: 27910729 DOI: 10.1080/00498254.2016.1217365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
1. The aim of this study was to identify the hepatic metabolic enzymes, which involved in the biotransformation of 6-benzyl-1-benzyloxymethyl-5-iodouracil (W-1), a novel non-nucleoside reverse transcriptase inhibitor (NNRTI) in rat and human in vitro. 2. The parent drug of W-1 was incubated with rat liver microsomes (RLMs) or recombinant CYPs (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4, and CYP3A5, respectively) in the presence or absence of nicotinamide adeninedinucleotide phosphate (NADPH)-regenerating system. The metabolites of W-1 were analyzed with liquid chromatography-ion trap-time of flight-mass spectrometry (LC-IT-TOF-MS). 3. The parent drug of W-1 was metabolized in a NADPH-dependent manner in RLMs. The kinetic parameters of prototype W-1 including Km, Vmax, and CLint were 2.3 μM, 3.3 nmol/min/mg protein, and 1.4 mL/min/mg protein, respectively. Two metabolites M1 and M2 were observed in shorter retention times (2.988 and 3.188 min) with a higher molecular ion at m/z 463.0160 (both M1 and M2) than that of the W-1 parent drug (6.158 min with m/z 447.0218). The CYP selective inhibition and recombinant enzymes also showed that two hydroxyl metabolites M1 and M2 are mainly mediated by CYP2C19 and CYP3A4. 4. The identification of CYPs involved in W-1 biotransformation is important to understand and minimize, if possible, the potential of drug-drug interactions.
Collapse
Affiliation(s)
- Ying-Yuan Lu
- a Department of Pharmacology , School of Basic Medical Science, Beijing (Peking) University , Beijing , PR China
| | - Hai-Xu Cheng
- a Department of Pharmacology , School of Basic Medical Science, Beijing (Peking) University , Beijing , PR China
| | - Xin Wang
- a Department of Pharmacology , School of Basic Medical Science, Beijing (Peking) University , Beijing , PR China
| | - Xiao-Wei Wang
- b Department of Chemical Biology , School of Pharmaceutical Sciences, Beijing (Peking) University , Beijing , PR China
| | - Jun-Yi Liu
- b Department of Chemical Biology , School of Pharmaceutical Sciences, Beijing (Peking) University , Beijing , PR China
| | - Pu Li
- a Department of Pharmacology , School of Basic Medical Science, Beijing (Peking) University , Beijing , PR China
| | - Ya-Qing Lou
- a Department of Pharmacology , School of Basic Medical Science, Beijing (Peking) University , Beijing , PR China
| | - Jun Li
- c State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Beijing (Peking) University , Beijing , PR China , and
| | - Chuang Lu
- d Department of Drug Metabolism & Pharmacokinetics , Biogen , Cambridge , MA , USA
| | - Guo-Liang Zhang
- a Department of Pharmacology , School of Basic Medical Science, Beijing (Peking) University , Beijing , PR China
| |
Collapse
|
58
|
Colbers A, Greupink R, Litjens C, Burger D, Russel FGM. Physiologically Based Modelling of Darunavir/Ritonavir Pharmacokinetics During Pregnancy. Clin Pharmacokinet 2016; 55:381-96. [PMID: 26369773 PMCID: PMC4761019 DOI: 10.1007/s40262-015-0325-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pregnant women are usually excluded from clinical trials. Physiologically based pharmacokinetic (PBPK) modelling may provide a method to predict pharmacokinetics in pregnant women, without the need to perform extensive in vivo clinical trials. Here, we used mechanistic modelling to delineate the potential impact of drug transporters on darunavir pharmacokinetics and to identify current knowledge gaps that limit accurate PBPK modelling of darunavir/ritonavir (darunavir/r) exposure in pregnancy. Simcyp (version 13.2) was used for PBPK modelling, using physicochemical and in vitro pharmacokinetic parameters of darunavir and ritonavir from the literature. The Michaelis-Menten constant (K m) and the maximum rate of metabolite formation (V max) for cytochrome P450 3A4-mediated darunavir biotransformation and inhibition by ritonavir were determined experimentally, while the contributions of hepatocyte influx and efflux transporters were assessed by sensitivity analysis. The simulations were compared with previously published clinical pharmacokinetic data. We found that use of a well-stirred liver model overestimated darunavir exposure substantially. A permeability-limited liver model, including hepatic uptake and efflux transporters and an efficient enterohepatic circulation step, resulted in an acceptable description of darunavir/r exposure. For the 600/100 mg darunavir/r twice-daily dose and the 800/100 mg once-daily dose, the estimated pharmacokinetic parameters were within a 2-fold range of the reported data. The predicted decreases in the area under the concentration-time curve (AUC) values during pregnancy for the twice- and once-daily doses were 27 and 41%, respectively, which were in line with the observed decreases of 17-22 and 33%. In conclusion, our data support a clinically relevant role of hepatic transporters in darunavir pharmacokinetics. By including them in our model, we successfully approximated the increase in darunavir exposure mediated by ritonavir co-administration and the decrease in darunavir exposure observed during pregnancy.
Collapse
Affiliation(s)
- Angela Colbers
- Department of Pharmacy, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Rick Greupink
- Department of Pharmacology and Toxicology (149), Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Carlijn Litjens
- Department of Pharmacy, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Department of Pharmacology and Toxicology (149), Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - David Burger
- Department of Pharmacy, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology (149), Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
59
|
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
|
60
|
Zetterberg C, Maltais F, Laitinen L, Liao S, Tsao H, Chakilam A, Hariparsad N. VX-509 (Decernotinib)-Mediated CYP3A Time-Dependent Inhibition: An Aldehyde Oxidase Metabolite as a Perpetrator of Drug-Drug Interactions. Drug Metab Dispos 2016; 44:1286-95. [PMID: 27298338 DOI: 10.1124/dmd.116.071100] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/10/2016] [Indexed: 02/13/2025] Open
Abstract
(R)-2-((2-(1H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-4-yl)amino)-2-methyl-N-(2,2,2-trifluoroethyl)butanamide (VX-509, decernotinib) is an oral Janus kinase 3 inhibitor that has been studied in patients with rheumatoid arthritis. Patients with rheumatoid arthritis often receive multiple medications, such as statins and steroids, to manage the signs and symptoms of comorbidities, which increases the chances of drug-drug interactions (DDIs). Mechanism-based inhibition is a subset of time-dependent inhibition (TDI) and occurs when a molecule forms a reactive metabolite which irreversibly binds and inactivates drug-metabolizing enzymes, potentially increasing the systemic load to toxic concentrations. Traditionally, perpetrating compounds are screened using human liver microsomes (HLMs); however, this system may be inadequate when the precipitant is activated by a non-cytochrome P450 (P450)-mediated pathway. Even though studies assessing competitive inhibition and TDI using HLM suggested a low risk for CYP3A4-mediated DDI in the clinic, VX-509 increased the area under the curve of midazolam, atorvastatin, and methyl-prednisolone by approximately 12.0-, 2.7-, and 4.3-fold, respectively. Metabolite identification studies using human liver cytosol indicated that VX-509 is converted to an oxidative metabolite, which is the perpetrator of the DDIs observed in the clinic. As opposed to HLM, hepatocytes contain the full complement of drug-metabolizing enzymes and transporters and can be used to assess TDI arising from non-P450-mediated metabolic pathways. In the current study, we highlight the role of aldehyde oxidase in the formation of the hydroxyl-metabolite of VX-509, which is involved in clinically significant TDI-based DDIs and represents an additional example in which a system-dependent prediction of TDI would be evident.
Collapse
Affiliation(s)
- Craig Zetterberg
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Francois Maltais
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Leena Laitinen
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Shengkai Liao
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Hong Tsao
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Ananthsrinivas Chakilam
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Niresh Hariparsad
- Drug Metabolism and Pharmacokinetics (C.Z., L.L., S.L., H.T., A.C., N.H.) and Department of Chemistry (F.M.), Vertex Pharmaceuticals Inc., Boston, Massachusetts
| |
Collapse
|
61
|
Yamazaki T, Desai A, Han D, Kato K, Kowalski D, Akhtar S, Lademacher C, Kovanda L, Townsend R. Pharmacokinetic Interaction Between Isavuconazole and a Fixed-Dose Combination of Lopinavir 400 mg/Ritonavir 100 mg in Healthy Subjects. Clin Pharmacol Drug Dev 2016; 6:93-101. [PMID: 27273248 PMCID: PMC5297880 DOI: 10.1002/cpdd.282] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/18/2016] [Accepted: 06/03/2016] [Indexed: 12/29/2022]
Abstract
This phase 1, open‐label study evaluated the pharmacokinetic effects of coadministration of the antifungal agent, isavuconazole (administered as its water‐soluble prodrug isavuconazonium sulfate), with the antiretroviral agent lopinavir/ritonavir in healthy adults. In part 1, 13 subjects were randomized to 2 arms to receive multiple doses of oral isavuconazole 100 mg either alone or with lopinavir/ritonavir 400/100 mg. In part 2, a different group of 55 subjects were randomized to 3 arms to receive multiple doses of oral isavuconazole 200 mg, either alone or with lopinavir/ritonavir 400/100 mg, or to receive oral lopinavir/ritonavir 400/100 mg alone. Mean area under the concentration‐time curve (AUC) following the last dose (AUCτ) and Cmax of isavuconazole increased by 113% and 96% in part 1 and by 96% and 74% in part 2 in the presence vs absence of lopinavir/ritonavir, respectively. Mean AUCτ and Cmax of lopinavir were 27% and 23% lower, and mean AUCτ and Cmax of ritonavir were 31% and 33% lower in the presence vs absence of isavuconazole, respectively. Mild to moderate gastrointestinal disorders were the most common adverse events experienced. These findings indicate that coadministration of lopinavir/ritonavir with isavuconazole can decrease the exposure of lopinavir/ritonavir and increase the exposure of isavuconazole. Patients should be monitored for reduced antiviral efficacy if these agents are coadministered.
Collapse
Affiliation(s)
- Takao Yamazaki
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | - Amit Desai
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | | | - Kota Kato
- Analysis & Pharmacokinetics Research Laboratories, Astellas Pharma Inc, Osaka, Japan
| | - Donna Kowalski
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | - Shahzad Akhtar
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | | | - Laura Kovanda
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | - Robert Townsend
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| |
Collapse
|
62
|
Mangold JB, Wu F, Rebello S. Compelling Relationship of CYP3A Induction to Levels of the Putative Biomarker 4β-Hydroxycholesterol and Changes in Midazolam Exposure. Clin Pharmacol Drug Dev 2016; 5:245-9. [DOI: 10.1002/cpdd.265] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/26/2016] [Indexed: 11/08/2022]
Affiliation(s)
- James B. Mangold
- Drug Metabolism and Pharmacokinetics; Novartis Institutes for Biomedical Research; East Hanover NJ USA
| | - Fan Wu
- Drug Metabolism and Pharmacokinetics; Novartis Institutes for Biomedical Research; East Hanover NJ USA
| | - Sam Rebello
- Drug Metabolism and Pharmacokinetics; Novartis Institutes for Biomedical Research; East Hanover NJ USA
| |
Collapse
|
63
|
Lebherz-Eichinger D, Tudor B, Krenn CG, Roth GA, Seemann R. Impact of different sedation protocols and perioperative procedures on patients admitted to the intensive care unit after maxillofacial tumor surgery of the lower jaw: A retrospective study. J Craniomaxillofac Surg 2016; 44:506-11. [PMID: 26874556 DOI: 10.1016/j.jcms.2015.12.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 11/25/2015] [Accepted: 12/30/2015] [Indexed: 10/22/2022] Open
Abstract
Maxillofacial tumor surgery often necessitates prolonged invasive ventilation to prevent blockage of the respiratory tract. To tolerate ventilation, continuously administered sedatives are recommended. Half-time of sedative or analgesic medication is an important characteristic by which narcotic drugs are chosen, due to the fact that weaning period increases with half-time. The aim of our study was to investigate whether a change in sedation regimen would affect the length of invasive ventilation or intensive care unit stay and medical costs. Additionally, the impact of various surgical procedures was analyzed. Data of 157 patients after mandibular surgery were retrospectively analyzed over 5 years in count regression models. Of those patients, 84 received a sedation regimen with sufentanil and midazolam and 73 with remifentanil and propofol. The impact of the surgical procedures (tracheostomy, tumor resection, neck dissection and length of operation) and the patient age and sex were analyzed with respect to length of ventilation and ICU days. Cost savings were calculated. Our data show that patients receiving remifentanil/propofol had fewer ventilation days (2.5 ± 2.5 versus 6.1 ± 4.6 days, P < 0.001) and were discharged earlier from the intensive care unit than patients receiving sufentanil/midazolam (5.1 ± 3.8 versus 9.2 ± 6.2 days, P < 0.001), leading to calculated cost savings of about 8000 Euro per patient. Length of operation negatively influenced length of ICU stay (P < 0.001). In conclusion, short-acting drugs such as remifentanil/propofol, as well as tracheostoma and shortened surgery duration may reduce the postoperative need for invasive ventilation and length of intensive care unit stay.
Collapse
Affiliation(s)
- Diana Lebherz-Eichinger
- Department of Anesthesiology, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria; RAIC Laboratory 13C1, Medical University of Vienna, Vienna, Austria
| | - Bianca Tudor
- Department of Anesthesiology, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria; RAIC Laboratory 13C1, Medical University of Vienna, Vienna, Austria
| | - Claus G Krenn
- Department of Anesthesiology, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria; RAIC Laboratory 13C1, Medical University of Vienna, Vienna, Austria
| | - Georg A Roth
- Department of Anesthesiology, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria; RAIC Laboratory 13C1, Medical University of Vienna, Vienna, Austria.
| | - Rudolf Seemann
- Department of Cranio-, Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
64
|
Greenblatt DJ. Evidence-based choice of ritonavir as index CYP3A inhibitor in drug-drug interaction studies. J Clin Pharmacol 2015; 56:152-6. [PMID: 26239522 DOI: 10.1002/jcph.609] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 07/31/2015] [Indexed: 12/23/2022]
Affiliation(s)
- David J Greenblatt
- Program in Pharmacology and Experimental Therapeutics, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
65
|
Martin P, Giardiello M, McDonald TO, Smith D, Siccardi M, Rannard SP, Owen A. Augmented Inhibition of CYP3A4 in Human Primary Hepatocytes by Ritonavir Solid Drug Nanoparticles. Mol Pharm 2015; 12:3556-68. [DOI: 10.1021/acs.molpharmaceut.5b00204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Philip Martin
- Department
of Molecular and Clinical Pharmacology, University of Liverpool, Block H, 70 Pembroke Place, Liverpool L69 3GF, U.K
| | - Marco Giardiello
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 3BX, U.K
| | - Tom O. McDonald
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 3BX, U.K
| | - Darren Smith
- Department
of Applied Sciences, University of Northumbria at Newcastle, Ellison
Building, Newcastle NE1
8ST, U.K
| | - Marco Siccardi
- Department
of Molecular and Clinical Pharmacology, University of Liverpool, Block H, 70 Pembroke Place, Liverpool L69 3GF, U.K
| | - Steven P. Rannard
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 3BX, U.K
| | - Andrew Owen
- Department
of Molecular and Clinical Pharmacology, University of Liverpool, Block H, 70 Pembroke Place, Liverpool L69 3GF, U.K
| |
Collapse
|
66
|
Kast RE, Karpel-Massler G, Halatsch ME. CUSP9* treatment protocol for recurrent glioblastoma: aprepitant, artesunate, auranofin, captopril, celecoxib, disulfiram, itraconazole, ritonavir, sertraline augmenting continuous low dose temozolomide. Oncotarget 2015; 5:8052-82. [PMID: 25211298 PMCID: PMC4226667 DOI: 10.18632/oncotarget.2408] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
CUSP9 treatment protocol for recurrent glioblastoma was published one year ago. We now present a slight modification, designated CUSP9*. CUSP9* drugs--aprepitant, artesunate, auranofin, captopril, celecoxib, disulfiram, itraconazole, sertraline, ritonavir, are all widely approved by regulatory authorities, marketed for non-cancer indications. Each drug inhibits one or more important growth-enhancing pathways used by glioblastoma. By blocking survival paths, the aim is to render temozolomide, the current standard cytotoxic drug used in primary glioblastoma treatment, more effective. Although esthetically unpleasing to use so many drugs at once, the closely similar drugs of the original CUSP9 used together have been well-tolerated when given on a compassionate-use basis in the cases that have come to our attention so far. We expect similarly good tolerability for CUSP9*. The combined action of this suite of drugs blocks signaling at, or the activity of, AKT phosphorylation, aldehyde dehydrogenase, angiotensin converting enzyme, carbonic anhydrase -2,- 9, -12, cyclooxygenase-1 and -2, cathepsin B, Hedgehog, interleukin-6, 5-lipoxygenase, matrix metalloproteinase -2 and -9, mammalian target of rapamycin, neurokinin-1, p-gp efflux pump, thioredoxin reductase, tissue factor, 20 kDa translationally controlled tumor protein, and vascular endothelial growth factor. We believe that given the current prognosis after a glioblastoma has recurred, a trial of CUSP9* is warranted.
Collapse
Affiliation(s)
| | - Georg Karpel-Massler
- University of Ulm, Department of Neurosurgery, Albert-Einstein-Allee 23, Ulm, Germany
| | - Marc-Eric Halatsch
- University of Ulm, Department of Neurosurgery, Albert-Einstein-Allee 23, Ulm, Germany
| |
Collapse
|
67
|
Greenblatt DJ, Harmatz JS. Ritonavir is the best alternative to ketoconazole as an index inhibitor of cytochrome P450-3A in drug-drug interaction studies. Br J Clin Pharmacol 2015; 80:342-50. [PMID: 25923589 DOI: 10.1111/bcp.12668] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/23/2015] [Accepted: 04/24/2015] [Indexed: 12/16/2022] Open
Abstract
AIMS The regulatory prohibition of ketoconazole as a CYP3A index inhibitor in drug-drug interaction (DDI) studies has compelled consideration of alternative inhibitors. METHODS The biomedical literature was searched to identify DDI studies in which oral midazolam (MDZ) was the victim, and the inhibitory perpetrator was either ketoconazole, itraconazole, clarithromycin, or ritonavir. The ratios (RAUC ) of total area under the curve (AUC) for MDZ with inhibitor divided by MDZ AUC in the control condition were aggregated across individual studies for each inhibitor. RESULTS Mean (± SE) RAUC values were: ketoconazole (15 studies, 131 subjects), 11.5 (±1.2); itraconazole (five studies, 48 subjects), 7.3 (±1.0); clarithromycin (five studies, 73 subjects), 6.5 (±10.9); and ritonavir (13 studies, 159 subjects), 14.5 (±2.0). Differences among inhibitors were significant (F = 5.31, P < 0.005). RAUC values were not significantly related to inhibitor dosage or to duration of inhibitor pre-exposure prior to administration of MDZ. CONCLUSIONS Ritonavir produces CYP3A inhibition equivalent to or greater than ketoconazole, and is the best index CYP3A inhibitor alternative to ketoconazole. Cobicistat closely resembles ritonavir in structure and function, and can also be considered. Itraconazole and clarithromycin are not suitable alternatives since they do not produce inhibition comparable with ketoconazole or ritonavir, and have other significant disadvantages as well.
Collapse
Affiliation(s)
- David J Greenblatt
- From the Program in Pharmacology and Experimental Therapeutics, Tufts University School of Medicine and Sackler School of Graduate Biomedical Sciences, Boston, MA, USA
| | - Jerold S Harmatz
- From the Program in Pharmacology and Experimental Therapeutics, Tufts University School of Medicine and Sackler School of Graduate Biomedical Sciences, Boston, MA, USA
| |
Collapse
|
68
|
Influence of HIV antiretrovirals on methadone N-demethylation and transport. Biochem Pharmacol 2015; 95:115-25. [PMID: 25801005 DOI: 10.1016/j.bcp.2015.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/12/2015] [Indexed: 02/02/2023]
Abstract
Drug interactions involving methadone and/or HIV antiretrovirals can be problematic. Mechanisms whereby antiretrovirals induce clinical methadone clearance are poorly understood. Methadone is N-demethylated to 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) by CYP2B6 and CYP3A4 in vitro, but by CYP2B6 in vivo. This investigation evaluated human hepatocytes as a model for methadone induction, and tested the hypothesis that methadone and EDDP are substrates for human drug transporters. Human hepatocyte induction by several antiretrovirals of methadone N-demethylation, and CYP2B6 and CYP3A4 transcription, protein expression and catalytic activity, and pregnane X receptor (PXR) activation were evaluated. Methadone and EDDP uptake and efflux by overexpressed transporters were also determined. Methadone N-demethylation was generally not significantly increased by the antiretrovirals. CYP2B6 mRNA and activity (bupropion N-demethylation) were induced by several antiretrovirals, as were CYP3A4 mRNA and protein expression, but only indinavir increased CYP3A activity (alfentanil dealkylation). CYP upregulation appeared related to PXR activation. Methadone was not a substrate for uptake (OCT1, OCT2, OCT3, OATP1A2, OATP1B1, OATP1B3, OATP2B1) or efflux (P-gp, BCRP) transporters. EDDP was a good substrate for P-gp, BCRP, OCT1, OCT3, OATP1A2, and OATP1B1. OATP1A2- and OCT3-mediated EDDP uptake, and BCRP-mediated EDDP efflux transport, was inhibited by several antiretrovirals. Results show that hepatocyte methadone N-demethylation resembles expressed and liver microsomal metabolism more than clinical metabolism. Compared with clinical studies, hepatocytes underreport induction of methadone metabolism by HIV drugs. Hepatocytes are not a good predictive model for clinical antiretroviral induction of methadone metabolism and not a substitute for clinical studies. EDDP is a transporter substrate, and is susceptible to transporter-mediated interactions.
Collapse
|
69
|
Khetani SR, Berger DR, Ballinger KR, Davidson MD, Lin C, Ware BR. Microengineered liver tissues for drug testing. ACTA ACUST UNITED AC 2015; 20:216-50. [PMID: 25617027 DOI: 10.1177/2211068214566939] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Indexed: 01/09/2023]
Abstract
Drug-induced liver injury (DILI) is a leading cause of drug attrition. Significant and well-documented differences between animals and humans in liver pathways now necessitate the use of human-relevant in vitro liver models for testing new chemical entities during preclinical drug development. Consequently, several human liver models with various levels of in vivo-like complexity have been developed for assessment of drug metabolism, toxicity, and efficacy on liver diseases. Recent trends leverage engineering tools, such as those adapted from the semiconductor industry, to enable precise control over the microenvironment of liver cells and to allow for miniaturization into formats amenable for higher throughput drug screening. Integration of liver models into organs-on-a-chip devices, permitting crosstalk between tissue types, is actively being pursued to obtain a systems-level understanding of drug effects. Here, we review the major trends, challenges, and opportunities associated with development and implementation of engineered liver models created from primary cells, cell lines, and stem cell-derived hepatocyte-like cells. We also present key applications where such models are currently making an impact and highlight areas for improvement. In the future, engineered liver models will prove useful for selecting drugs that are efficacious, safer, and, in some cases, personalized for specific patient populations.
Collapse
Affiliation(s)
- Salman R Khetani
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Dustin R Berger
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Kimberly R Ballinger
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Matthew D Davidson
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Christine Lin
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Brenton R Ware
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
70
|
Abstract
Hepatocytes in sandwich configuration constitute of primary hepatocytes cultured between two layers of extracellular matrix. Sandwich-cultured hepatocytes maintain expression of liver-specific proteins and gradually form intact bile canaliculi with functional biliary excretion of endogenous compounds and xenobiotics. Both freshly isolated and cryopreserved hepatocytes can be used to establish sandwich cultures. Therefore, this preclinical model has become an invaluable in vitro tool to evaluate hepatobiliary drug transport, metabolism, hepatotoxicity, and drug interactions. In this chapter, commonly used procedures to cultivate primary hepatocytes from human and rat in sandwich configuration are described.
Collapse
Affiliation(s)
- Janneke Keemink
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, O&N2, Herestraat 49, Bus 921, Leuven, 3000, Belgium
| | | | | |
Collapse
|
71
|
Loue C, Tod M. Reliability and extension of quantitative prediction of CYP3A4-mediated drug interactions based on clinical data. AAPS JOURNAL 2014; 16:1309-20. [PMID: 25274605 DOI: 10.1208/s12248-014-9663-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/02/2014] [Indexed: 01/06/2023]
Abstract
An approach was proposed in 2007 for quantitative predictions of cytochrome P450 (CYP)3A4-mediated drug-drug interactions. It is based on two characteristic parameters: the contribution ratio (CR; i.e., the fraction of victim drug clearance by CYP) and the inhibition ratio (IR) of the inhibitor. Knowledge of these parameters allows forecasting of the ratio between the area under the plasma concentration-time curve (AUC) of the victim drug when given with the inhibitor and the AUC of the victim drug when it is given alone. So far, these parameters were established for 21 substrates and 17 inhibitors. The goals of our study were to test the assumption of substrate independence of the potency of inhibitors in vivo and to estimate the CR and IR for an extended list of substrates and inhibitors of CYP3A4. The assumption of independence of IRs from the substrate was evaluated on a set of eight victim drugs and eight inhibitors. Forty-four AUC ratios were available. This assumption was rejected in four cases, but it did not result in more than a twofold error in AUC ratio predictions. The extended list of substrates and inhibitors was defined by a thorough literature search. Fifty-nine AUC ratios were available for the global analysis. Final estimates of CRs and IRs were obtained for 37 substrates and 25 inhibitors, respectively. The mean prediction error of the ratios was 0.02, while the mean absolute prediction error was 0.58. Predictive distributions for 917 possible interactions were obtained, giving detailed information on some drugs or inhibitors that have been poorly studied so far.
Collapse
Affiliation(s)
- Constance Loue
- Pharmacie, Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France
| | | |
Collapse
|
72
|
Zhang JG, Ho T, Callendrello AL, Clark RJ, Santone EA, Kinsman S, Xiao D, Fox LG, Einolf HJ, Stresser DM. Evaluation of calibration curve-based approaches to predict clinical inducers and noninducers of CYP3A4 with plated human hepatocytes. Drug Metab Dispos 2014; 42:1379-91. [PMID: 24924386 DOI: 10.1124/dmd.114.058602] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Cytochrome P450 (P450) induction is often considered a liability in drug development. Using calibration curve-based approaches, we assessed the induction parameters R3 (a term indicating the amount of P450 induction in the liver, expressed as a ratio between 0 and 1), relative induction score, Cmax/EC50, and area under the curve (AUC)/F2 (the concentration causing 2-fold increase from baseline of the dose-response curve), derived from concentration-response curves of CYP3A4 mRNA and enzyme activity data in vitro, as predictors of CYP3A4 induction potential in vivo. Plated cryopreserved human hepatocytes from three donors were treated with 20 test compounds, including several clinical inducers and noninducers of CYP3A4. After the 2-day treatment, CYP3A4 mRNA levels and testosterone 6β-hydroxylase activity were determined by real-time reverse transcription polymerase chain reaction and liquid chromatography-tandem mass spectrometry analysis, respectively. Our results demonstrated a strong and predictive relationship between the extent of midazolam AUC change in humans and the various parameters calculated from both CYP3A4 mRNA and enzyme activity. The relationships exhibited with non-midazolam in vivo probes, in aggregate, were unsatisfactory. In general, the models yielded better fits when unbound rather than total plasma Cmax was used to calculate the induction parameters, as evidenced by higher R(2) and lower root mean square error (RMSE) and geometric mean fold error. With midazolam, the R3 cut-off value of 0.9, as suggested by US Food and Drug Administration guidance, effectively categorized strong inducers but was less effective in classifying midrange or weak inducers. This study supports the use of calibration curves generated from in vitro mRNA induction response curves to predict CYP3A4 induction potential in human. With the caveat that most compounds evaluated here were not strong inhibitors of enzyme activity, testosterone 6β-hydroxylase activity was also demonstrated to be a strong predictor of CYP3A4 induction potential in this assay model.
Collapse
Affiliation(s)
- J George Zhang
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Thuy Ho
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Alanna L Callendrello
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Robert J Clark
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Elizabeth A Santone
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Sarah Kinsman
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Deqing Xiao
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Lisa G Fox
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - Heidi J Einolf
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| | - David M Stresser
- Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (J.G.Z., T.H., A.L.C., R.J.C., E.A.S., S.K., D.X., L.G.F., D.M.S.); and Novartis Institutes for Biomedical Research, East Hanover, New Jersey (H.J.E.)
| |
Collapse
|
73
|
DuBois BN, Atrio J, Stanczyk FZ, Cherala G. Increased exposure of norethindrone in HIV+ women treated with ritonavir-boosted atazanavir therapy. Contraception 2014; 91:71-5. [PMID: 25245190 DOI: 10.1016/j.contraception.2014.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/04/2014] [Accepted: 08/16/2014] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Pharmacokinetics of norethindrone in combination oral contraceptive regimen are well described among HIV+ women treated with ritonavir-boosted protease inhibitor therapies; however, such characterization is lacking in women using progestin-only contraception. Our objective is to characterize pharmacokinetics of norethindrone in HIV+ women using ritonavir-boosted atazanavir treatment during progestin-only contraceptive regimens. STUDY DESIGN An open-label, prospective, nonrandomized trial to characterize the pharmacokinetics of norethindrone in HIV+ women receiving ritonavir-boosted atazanavir (n=10; treatment group) and other antiretroviral therapy known to not alter norethindrone levels (n=17; control group) was conducted. Following informed consent, women were instructed to take a single daily fixed oral dose of 0.35 mg norethindrone and 300 mg/100 mg atazanavir/ritonavir for 22 days. On day 22, serial blood samples were collected by venous catheter at 0, 1, 2, 3, 4, 6, 8, 12, 24, 48 and 72 h. Whole blood was processed to collect serum and stored at -20°C until later analysis using radioimmunoassay. Pharmacokinetic parameters were estimated using noncompartmental method. RESULTS In the treatment group, compared to the control group, an increase in area under the curve₀₋₂₄ (16.69 h*ng/mL vs. 25.20 h*ng/mL; p<.05) and maximum serum concentration (2.09 ng/mL vs. 3.19 ng/mL; p<.05), decrease (25%-40%) in apparent volume of distribution and apparent clearance, and unaltered half-life were observed. CONCLUSION(S) Our findings suggest that progestin-only contraceptives, unlike combination oral contraceptives, benefit from drug-drug interaction and achieve higher levels of exposure. Further studies are needed to establish whether pharmacokinetic interaction leads to favorable clinical outcomes. IMPLICATIONS Norethindrone-based progestin-only contraceptives, unlike combination oral contraceptives, exhibit greater drug exposure when co-administered with ritonavir-boosted atazanavir regimen and thus may not warrant a category 3 designation by the World Health Organization. Prospective studies are needed to confirm whether pharmacokinetic interaction results in favorable clinical outcomes.
Collapse
Affiliation(s)
- Barent N DuBois
- Oregon State University & Oregon Health Science University College of Pharmacy, Portland, OR, USA
| | - Jessica Atrio
- Department of Obstetrics and Gynecology, Albert Einstein College of Medicine, New York, NY, USA
| | - Frank Z Stanczyk
- Department of Obstetrics and Gynecology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Ganesh Cherala
- Oregon State University & Oregon Health Science University College of Pharmacy, Portland, OR, USA; Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
74
|
Kaspera R, Kirby BJ, Sahele T, Collier AC, Kharasch ED, Unadkat JD, Totah RA. Investigating the contribution of CYP2J2 to ritonavir metabolism in vitro and in vivo. Biochem Pharmacol 2014; 91:109-18. [PMID: 24973543 DOI: 10.1016/j.bcp.2014.06.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 10/25/2022]
Abstract
Ritonavir, an HIV protease inhibitor, is successfully used for the prevention and treatment of HIV infections. Ritonavir pharmacokinetics are complicated by inhibition, induction and pharmacogenetics of cytochrome P450 (CYP) enzymes mediating its clearance. This investigation revealed that CYP2J2, along with CYP3A4/5 and CYP2D6, efficiently metabolizes ritonavir, and to a CYP2J2-specific (minor) metabolite. Chemical inhibition of ritonavir metabolism, clearance, KI/kinact and abundance of CYP2J2 in liver microsomes were evaluated and then applied to an in vitro-in vivo static scaling model to estimate the contribution of each isozyme, as a function of CYP abundance, activity, and genotype. Disposition of the CYP2J2-specific metabolite was also evaluated in vivo. In plasma, metabolite abundance was well above previously reported levels with circulating concentrations measured at 2 μM for the main hydroxylisopropyl metabolite. Ritonavir and metabolite plasma profiles were simulated using Simcyp(®). A modest (2-6%) contribution of CYP2J2 to ritonavir clearance is predicted which increases to more than 20% in subjects carrying CYP2D6 poor metabolizer polymorphisms and CYP3A4 irreversible inhibition. These results indicate that minor drug metabolizing enzymes could become quantitatively important in RTV clearance if main metabolic pathways are impeded.
Collapse
Affiliation(s)
- Rüdiger Kaspera
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195-7610, USA.
| | - Brian J Kirby
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357630, Seattle, WA 98195-7630, USA.
| | - Tariku Sahele
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195-7610, USA.
| | - Ann C Collier
- Department of Medicine, University of Washington, Box 359929, Seattle, WA 98195-9929, USA.
| | - Evan D Kharasch
- Department of Anesthesiology, School of Medicine, Washington University, 660 South Euclid Avenue, Campus Box 8054, St. Louis, MO 63110, USA.
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Box 357630, Seattle, WA 98195-7630, USA.
| | - Rheem A Totah
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Box 357610, Seattle, WA 98195-7610, USA.
| |
Collapse
|
75
|
Phase I safety, pharmacokinetics, and pharmacogenetics study of the antituberculosis drug PA-824 with concomitant lopinavir-ritonavir, efavirenz, or rifampin. Antimicrob Agents Chemother 2014; 58:5245-52. [PMID: 24957823 DOI: 10.1128/aac.03332-14] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
There is an urgent need for new antituberculosis (anti-TB) drugs, including agents that are safe and effective with concomitant antiretrovirals (ARV) and first-line TB drugs. PA-824 is a novel antituberculosis nitroimidazole in late-phase clinical development. Cytochrome P450 (CYP) 3A, which can be induced or inhibited by ARV and antituberculosis drugs, is a minor (∼20%) metabolic pathway for PA-824. In a phase I clinical trial, we characterized interactions between PA-824 and efavirenz (arm 1), lopinavir/ritonavir (arm 2), and rifampin (arm 3) in healthy, HIV-uninfected volunteers without TB disease. Participants in arms 1 and 2 were randomized to receive drugs via sequence 1 (PA-824 alone, washout, ARV, and ARV plus PA-824) or sequence 2 (ARV, ARV with PA-824, washout, and PA-824 alone). In arm 3, participants received PA-824 and then rifampin and then both. Pharmacokinetic sampling occurred at the end of each dosing period. Fifty-two individuals participated. Compared to PA-824 alone, plasma PA-824 values (based on geometric mean ratios) for maximum concentration (Cmax), area under the concentration-time curve from 0 to 24 h (AUC0-24), and trough concentration (Cmin) were reduced 28%, 35%, and 46% with efavirenz, 13%, 17%, and 21% with lopinavir-ritonavir (lopinavir/r) and 53%, 66%, and 85% with rifampin, respectively. Medications were well tolerated. In conclusion, lopinavir/r had minimal effect on PA-824 exposures, supporting PA-824 use with lopinavir/r without dose adjustment. PA-824 exposures, though, were reduced more than expected when given with efavirenz or rifampin. The clinical implications of these reductions will depend upon data from current clinical trials defining PA-824 concentration-effect relationships. (This study has been registered at ClinicalTrials.gov under registration no. NCT01571414.).
Collapse
|
76
|
Siccardi M, Marzolini C, Seden K, Almond L, Kirov A, Khoo S, Owen A, Back D. Prediction of drug-drug interactions between various antidepressants and efavirenz or boosted protease inhibitors using a physiologically based pharmacokinetic modelling approach. Clin Pharmacokinet 2014; 52:583-92. [PMID: 23479398 DOI: 10.1007/s40262-013-0056-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVE The rate of depression in patients with HIV is higher than in the general population. The use of antidepressants can have a beneficial effect, improving antiretroviral therapy adherence and consequently their efficacy and safety. Efavirenz and protease inhibitor boosted with ritonavir are major components of the antiretroviral therapy and are inducers and/or inhibitors of several cytochrome P450 (CYP) isoforms. Although antidepressants are prescribed to a significant proportion of patients treated with antiretrovirals, there are limited clinical data on drug-drug interactions. The aim of this study was to predict the magnitude of drug-drug interactions among efavirenz, boosted protease inhibitors and the most commonly prescribed antidepressants using an in vitro-in vivo extrapolation (IVIVE) model simulating virtual clinical trials. METHODS In vitro data describing the chemical characteristics, and absorption, distribution, metabolism and elimination (ADME) properties of efavirenz, boosted protease inhibitors and the most commonly prescribed antidepressants were obtained from published literature or generated by standard methods. Pharmacokinetics and drug-drug interaction were simulated using the full physiologically based pharmacokinetic model implemented in the Simcyp™ ADME simulator. The robustness of our modeling approach was assessed by comparing the magnitude of simulated drug-drug interactions using probe drugs to that observed in clinical studies. RESULTS Simulated pharmacokinetics and drug-drug interactions were in concordance with available clinical data. Although the simulated drug-drug interactions with antidepressants were overall weak to moderate according to the classification of the US FDA, fluoxetine and venlafaxine represent better candidates from a pharmacokinetic standpoint for patients on efavirenz and venlafaxine or citalopram for patients on boosted protease inhibitors. CONCLUSION The modest magnitude of interaction could be explained by the fact that antidepressants are substrates of multiple isoforms and thus metabolism can still occur through CYPs that are weakly impacted by efavirenz or boosted protease inhibitors. These findings indicate that IVIVE is a useful tool for predicting drug-drug interactions and designing prospective clinical trials, giving insight into the variability of exposure, sample size and time-dependent induction or inhibition.
Collapse
Affiliation(s)
- Marco Siccardi
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, 70 Pembroke Place, Liverpool, L69 3GF, UK.
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Ring B, Wrighton SA, Mohutsky M. Reversible mechanisms of enzyme inhibition and resulting clinical significance. Methods Mol Biol 2014; 1113:37-56. [PMID: 24523108 DOI: 10.1007/978-1-62703-758-7_4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Inhibition of a drug-metabolizing enzyme by the reversible interaction of a drug with the enzyme, thus decreasing the metabolism of another drug, is a major cause of clinically significant drug-drug interactions. This chapter defines the four reversible mechanisms of inhibition exhibited by drugs: competitive, noncompetitive, uncompetitive, and mixed competitive/noncompetitive. An in vitro procedure to determine the potential of a drug to be a reversible inhibitor is also provided. Finally, a number of examples of clinically significant drug-drug interactions resulting from reversible inhibition are described.
Collapse
Affiliation(s)
- Barbara Ring
- Quintiles, 5225 Exploration Drive, Indianapolis, IN, 46241, USA
| | | | | |
Collapse
|
78
|
Kharasch ED, Stubbert K. Cytochrome P4503A does not mediate the interaction between methadone and ritonavir-lopinavir. Drug Metab Dispos 2013; 41:2166-74. [PMID: 24067429 PMCID: PMC3834136 DOI: 10.1124/dmd.113.053991] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/25/2013] [Indexed: 01/12/2023] Open
Abstract
Plasma concentrations of orally administered methadone are reduced by the human immunodeficiency virus protease inhibitor combination ritonavir and lopinavir, but the mechanism is unknown. Methadone metabolism, clearance, and drug interactions have been attributed to CYP3A4, but this remains controversial. This investigation assessed the effects of acute (2 days) and steady-state (2 weeks) ritonavir-lopinavir on intravenous and oral methadone metabolism and clearance, hepatic and intestinal CYP3A4/5 activity (using the probe substrate intravenous and oral alfentanil), and intestinal transporter activity (using oral fexofenadine) in healthy volunteers. Plasma and urine concentrations of methadone and metabolite enantiomers, and other analytes, were determined by mass spectrometry. Acute and chronic ritonavir-lopinavir reduced plasma methadone enantiomer concentrations in half, with an average 2.6- and 1.5-fold induction of systemic and apparent oral methadone clearances. Induction was attributable to stereoselectively increased hepatic methadone N-demethylation, hepatic extraction, and hepatic clearance, and there was a strong correlation between methadone N-demethylation and clearance. Methadone renal clearance was unchanged. Alfentanil's systemic clearance and hepatic extraction, apparent oral clearance, and intestinal extraction were reduced to 25%, 16%, and 35% of control, indicating strong inhibition of hepatic and intestinal CYP3A activities. Ritonavir-lopinavir (acute > chronic) increased fexofenadine exposure, suggesting intestinal P-glycoprotein inhibition. No correlation was found between methadone clearance and CYP3A activity. Acute and steady-state ritonavir-lopinavir stereoselectively induced methadone N-demethylation and clearance, despite significant inhibition of hepatic and intestinal CYP3A activity. Ritonavir-lopinavir inhibited intestinal transporters activity but had no effect on methadone bioavailability. These results do not support a significant role for CYP3A or ritonavir-lopinavir-inhibitable intestinal transporters in single-dose methadone disposition.
Collapse
Affiliation(s)
- Evan D Kharasch
- Department of Anesthesiology, Division of Clinical and Translational Research (E.D.K., K.S.), and Department of Biochemistry and Molecular Biophysics (E.D.K.), Washington University in St. Louis, St. Louis, Missouri
| | | |
Collapse
|
79
|
Lin HL, D'Agostino J, Kenaan C, Calinski D, Hollenberg PF. The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Drug Metab Dispos 2013; 41:1813-24. [PMID: 23886699 PMCID: PMC3781371 DOI: 10.1124/dmd.113.053108] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/25/2013] [Indexed: 12/28/2022] Open
Abstract
The mechanism-based inactivation of human CYP2B6 by ritonavir (RTV) in a reconstituted system was investigated. The inactivation is time, concentration, and NADPH dependent and exhibits a K(I) of 0.9 μM, a k(inact) of 0.05 min⁻¹, and a partition ratio of approximately 3. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis showed that the protonated molecular ion of RTV exhibits an m/z at 721 and its two major metabolites are an oxidation product with MH⁺ at m/z 737 and a deacylated product with MH⁺ at m/z 580. Inactivation of CYP2B6 by incubation with 10 μM RTV for 10 min resulted in an approximately 50% loss of catalytic activity and native heme, but no modification of the apoprotein was observed. RTV was found to be a potent mixed-type reversible inhibitor (K(i) = 0.33 μM) and a type II ligand (spectral dissociation constant-K(s) = 0.85 μM) of CYP2B6. Although previous studies have demonstrated that RTV is a potent mechanism-based inactivator of CYP3A4, the molecular mechanism responsible for the inactivation has not been determined. Here, we provide evidence that RTV inactivation of CYP3A4 is due to heme destruction with the formation of a heme-protein adduct. Similar to CYP2B6, there is no significant modification of the apoprotein. Furthermore, LC-MS/MS analysis revealed that both CYP3A4 and human liver microsomes form an RTV-glutathione conjugate having a MH⁺ at m/z 858 during metabolism of RTV, suggesting the formation of an isocyanate intermediate leading to formation of the conjugate.
Collapse
Affiliation(s)
- Hsia-lien Lin
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | |
Collapse
|
80
|
Tod M, Nkoud-Mongo C, Gueyffier F. Impact of genetic polymorphism on drug-drug interactions mediated by cytochromes: a general approach. AAPS JOURNAL 2013; 15:1242-52. [PMID: 24027036 DOI: 10.1208/s12248-013-9530-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 08/19/2013] [Indexed: 11/30/2022]
Abstract
Currently, quantitative prediction of the impact of genetic polymorphism and drug-drug interactions mediated by cytochromes, based on in vivo data, is made by two separate methods and restricted to a single cytochrome. We propose a unified approach for describing the combined impact of drug-drug interactions and genetic polymorphism on drug exposure. It relies on in vivo data and uses the following three characteristic parameters: one for the victim drug, one for the interacting drug, and another for the genotype. These parameters are known for a wide range of drugs and genotypes. The metrics of interest are the ratio of victim drug area under the curve (AUC) in patients with genetic variants taking both drugs, to the AUC in patients with either variant or wild-type genotype taking the victim drug alone. The approach was evaluated by external validation, comparing predicted and observed AUC ratios found in the literature. Data were found for 22 substrates, 30 interacting drugs, and 38 substrate-interacting drug couples. The mean prediction error of AUC ratios was 0.02, and the mean prediction absolute error was 0.38 and 1.34, respectively. The model may be used to predict the variations in exposure resulting from a number of drug-drug-genotype combinations. The proposed approach will help (1) to identify comedications and population at risk, (2) to adapt dosing regimens, and (3) to prioritize the clinical pharmacokinetic studies to be done.
Collapse
Affiliation(s)
- Michel Tod
- Hospices Civils de Lyon, Université de Lyon, Université Lyon 1, 69000, Lyon, France,
| | | | | |
Collapse
|
81
|
Berry LM, Zhao Z, Lin MHJ. Dynamic modeling of cytochrome P450 inhibition in vitro: impact of inhibitor depletion on IC₅₀ shift. Drug Metab Dispos 2013; 41:1433-41. [PMID: 23649703 DOI: 10.1124/dmd.113.051508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The impact of inhibitor depletion on the determination of shifted IC₅₀ (IC₅₀ determined after 30 minutes of preincubation with inhibitor) is examined. In addition, IC₅₀-shift data are analyzed using a mechanistic model that incorporates the processes of inhibitor depletion, as well as reversible and time-dependent inhibition. Anomalies such as a smaller-than-expected shift in IC₅₀ and even increases in IC₅₀ with preincubation were explained by the depletion of inhibitor during the preincubation. The IC₅₀-shift assay remains a viable approach to characterizing a wide range of reversible and time-dependent inhibitors. However, as with more traditional time-dependent inactivation methods, it is recommended that IC₅₀-shift experimental data be interpreted with some knowledge of the magnitude of inhibitor depletion. For the most realistic classification of time-dependent inhibitors using IC₅₀-shift methods, shifted IC₅₀ should be calculated using observed inhibitor concentrations at the end of the incubation rather than nominal inhibitor concentrations. Finally, a mechanistic model that includes key processes, such as competitive inhibition, enzyme inactivation, and inhibitor depletion, can be used to describe accurately the observed IC₅₀ and shifted IC₅₀ curves. For compounds showing an IC₅₀ fold shift >1.5 based on the observed inhibitor concentrations, reanalyzing the IC₅₀-shift data using the mechanistic model appeared to allow for reasonable estimation of Ki, KI, and kinact directly from the IC₅₀ shift experiments.
Collapse
Affiliation(s)
- Loren M Berry
- Pharmacokinetics and Drug Metabolism, Amgen, Inc., 360 Binney St., Cambridge, MA 02142, USA.
| | | | | |
Collapse
|
82
|
Pharmacokinetics of phase I nevirapine metabolites following a single dose and at steady state. Antimicrob Agents Chemother 2013; 57:2154-60. [PMID: 23459477 DOI: 10.1128/aac.02294-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nevirapine is one of the most extensively prescribed antiretrovirals worldwide. The present analyses used data and specimens from two prior studies to characterize and compare plasma nevirapine phase I metabolite profiles following a single 200-mg oral dose of nevirapine in 10 HIV-negative African Americans and a steady-state 200-mg twice-daily dose in 10 HIV-infected Cambodians. Nevirapine was assayed by high-performance liquid chromatography (HPLC). The 2-, 3-, 8- and 12-hydroxy and 4-carboxy metabolites of nevirapine were assayed by liquid chromatography-tandem mass spectrometry (LC/MS/MS). Pharmacokinetic parameters were calculated by noncompartmental analysis. The metabolic index for each metabolite was defined as the ratio of the metabolite area under the concentration-time curve (AUC) to the nevirapine AUC. Every metabolite concentration was much less than the corresponding nevirapine concentration. The predominant metabolite after single dose and at steady state was 12-hydroxynevirapine. From single dose to steady state, the metabolic index increased for 3-hydroxynevirapine (P < 0.01) but decreased for 2-hydroxynevirapine (P < 0.001). The 3-hydroxynevirapine metabolic index was correlated with nevirapine apparent clearance (P < 0.001). These findings are consistent with induction of CYP2B6 (3-hydroxy metabolite) and a possible inhibition of CYP3A (2-hydroxy metabolite), although these are preliminary data. There were no such changes in metabolic indexes for 12-hydroxynevirapine or 4-carboxynevirapine. Two subjects with the CYP2B6 *6*6 genetic polymorphism had metabolic indexes in the same range as other subjects. These results suggest that nevirapine metabolite profiles change over time under the influence of enzyme induction, enzyme inhibition, and host genetics. Further work is warranted to elucidate nevirapine biotransformation pathways and implications for drug efficacy and toxicity.
Collapse
|
83
|
De Bruyn T, Chatterjee S, Fattah S, Keemink J, Nicolaï J, Augustijns P, Annaert P. Sandwich-cultured hepatocytes: utility for in vitro exploration of hepatobiliary drug disposition and drug-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2013; 9:589-616. [PMID: 23452081 DOI: 10.1517/17425255.2013.773973] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The sandwich-cultured hepatocyte (SCH) model has become an invaluable in vitro tool for studying hepatic drug transport, metabolism, biliary excretion and toxicity. The relevant expression of many hepatocyte-specific functions together with the in vivo-like morphology favor SCHs over other preclinical models for evaluating hepatobiliary drug disposition and drug-induced hepatotoxicity. AREAS COVERED In this review, the authors highlight recommended procedures required for reproducibly culturing hepatocytes in sandwich configuration. It also provides an overview of the SCH model characteristics as a function of culture time. Lastly, the article presents a summary of the most prominent applications of the SCH model, including hepatic drug clearance prediction, drug-drug interaction potential and drug-induced hepatotoxicity. EXPERT OPINION When human (cryopreserved) hepatocytes are used to establish sandwich cultures, the model appears particularly valuable to quantitatively investigate clinically relevant mechanisms related to in vivo hepatobiliary drug disposition and hepatotoxicity. Nonetheless, the SCH model would largely benefit from better insight into the fundamental cell signaling mechanisms that are critical for long-term in vitro maintenance of the hepatocytic phenotype. Studies systematically exploring improved cell culture conditions (e.g., co-cultures or extracellular matrix modifications), as well as in vitro work identifying key transcription factors involved in hepatocyte differentiation are currently emerging.
Collapse
Affiliation(s)
- Tom De Bruyn
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, O&N2, Herestraat 49-bus-921, 3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
84
|
Mao J, Johnson TR, Shen Z, Yamazaki S. Prediction of crizotinib-midazolam interaction using the Simcyp population-based simulator: comparison of CYP3A time-dependent inhibition between human liver microsomes versus hepatocytes. Drug Metab Dispos 2013; 41:343-52. [PMID: 23129213 DOI: 10.1124/dmd.112.049114] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Crizotinib (Xalkori) is an orally available potent inhibitor of multiple tyrosine kinases, including anaplastic lymphoma kinase and mesenchymal-epithelial transition factor. Objectives of the present study were as follows: 1) to characterize crizotinib time-dependent inhibition (TDI) potency for CYP3A in human liver microsomes (HLM) and cryopreserved human hepatocytes suspended in human plasma (HSP); 2) to characterize crizotinib enzyme induction potency on CYP3A4 in cryopreserved human hepatocytes; 3) to predict crizotinib steady-state plasma concentrations in patients (e.g., autoinhibition and autoinduction) using the mechanistic dynamic model, Simcyp population-based simulator; and 4) to predict a clinical crizotinib-midazolam interaction using the dynamic model as well as the static mathematical model. Crizotinib inactivation constant (K(I)) and maximum inactivation rate constant (k(inact)) for TDI were estimated as, respectively, 0.37 µM and 6.9 h(-1) in HLM and 0.89 µM and 0.78 h(-1) in HSP. Thus, crizotinib inactivation efficiency (k(inact)/K(I)) was ∼20-fold lower in HSP relative to HLM. Crizotinib E(max) and EC(50) for CYP3A4 induction (measured as mRNA expression) were estimated as 6.4- to 29-fold and 0.47 to 3.1 µM, respectively. Based on these in vitro parameters, the predicted crizotinib steady-state area under plasma concentration-time curve (AUC) with HLM-TDI was 2.1-fold higher than the observed AUC, whereas that with HSP-TDI was consistent with the observed result (≤1.1-fold). The increase in midazolam AUC with coadministration of crizotinib (21-fold) was significantly overpredicted using HLM-TDI, whereas the prediction using HSP-TDI (3.6-fold) was consistent with the observed result (3.7-fold). Collectively, the present study demonstrated the value of HSP to predict in vivo CYP3A-mediated drug-drug interaction.
Collapse
Affiliation(s)
- Jialin Mao
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, San Diego, CA, USA
| | | | | | | |
Collapse
|
85
|
Aasa J, Hu Y, Eklund G, Lindgren A, Baranczewski P, Malmquist J, Turek D, Bueters T. Effect of solvents on the time-dependent inhibition of CYP3A4 and the biotransformation of AZD3839 in human liver microsomes and hepatocytes. Drug Metab Dispos 2013; 41:159-69. [PMID: 23073735 DOI: 10.1124/dmd.112.047597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Time-dependent inhibition (TDI) of the cytochrome P450 (P450) family of enzymes is usually studied in human liver microsomes (HLM) by investigating whether the inhibitory potency is increased with increased incubation times. The presented work was initiated after a discrepancy was observed for the TDI of an important P450 enzyme, CYP3A4, during early studies of the investigational drug compound AZD3839 [(S)-1-(2-(difluoromethyl)pyridin-4-yl)-4-fluoro-1-(3-(pyrimidin-5-yl)phenyl)-1H-isoindol-3-amine hemifumarate]; TDI was detected using a regulatory method but not with an early screening method. We show here that the different solvents present in the respective studies, dimethyl sulfoxide (DMSO, screening method) versus methanol or water (regulatory method), were responsible for the different TDI results. We further demonstrate why DMSO, present at the levels of 0.2% and 0.5% in the incubations, masked the TDI effect. In addition to the TDI experiments performed in HLM, TDI studies with AZD3839 were performed in pooled human hepatocytes (Hhep) from different suppliers, using DMSO, methanol, or water. The results from these experiments show no TDI or attenuated TDI effect, depending on the supplier. Metabolite identification of the compound dissolved in DMSO, methanol, or water shows different profiles after incubations with the different systems (HLM or Hhep), which may explain the differences in the TDI outcomes. Thorough investigations of the biotransformation of AZD3839 have been performed to find the reactive pathway causing the TDI of CYP3A4, and are presented here. Our findings show that the in vitro risk profile for drug-drug interactions potential of AZD3839 is very much dependent on the chosen test system and the experimental conditions used.
Collapse
Affiliation(s)
- Jenny Aasa
- DMPK CNSP iMed Science, AstraZeneca R&D, Innovative Medicines, SE-15185, Södertälje, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
86
|
A PBPK Model to Predict Disposition of CYP3A-Metabolized Drugs in Pregnant Women: Verification and Discerning the Site of CYP3A Induction. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2012; 1:e3. [PMID: 23835883 PMCID: PMC3606941 DOI: 10.1038/psp.2012.2] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Besides logistical and ethical concerns, evaluation of safety and efficacy of medications in pregnant women is complicated by marked changes in pharmacokinetics (PK) of drugs. For example, CYP3A activity is induced during the third trimester (T3). We explored whether a previously published physiologically based pharmacokinetic (PBPK) model could quantitatively predict PK profiles of CYP3A-metabolized drugs during T3, and discern the site of CYP3A induction (i.e., liver, intestine, or both). The model accounted for gestational age-dependent changes in maternal physiological function and hepatic CYP3A activity. For model verification, mean plasma area under the curve (AUC), peak plasma concentration (Cmax), and trough plasma concentration (Cmin) of midazolam (MDZ), nifedipine (NIF), and indinavir (IDV) were predicted and compared with published studies. The PBPK model successfully predicted MDZ, NIF, and IDV disposition during T3. A sensitivity analysis suggested that CYP3A induction in T3 is most likely hepatic and not intestinal. Our PBPK model is a useful tool to evaluate different dosing regimens during T3 for drugs cleared primarily via CYP3A metabolism.
Collapse
|
87
|
Liu L, Mugundu GM, Kirby BJ, Samineni D, Desai PB, Unadkat JD. Quantification of human hepatocyte cytochrome P450 enzymes and transporters induced by HIV protease inhibitors using newly validated LC-MS/MS cocktail assays and RT-PCR. Biopharm Drug Dispos 2012; 33:207-17. [PMID: 22498895 DOI: 10.1002/bdd.1788] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 04/10/2012] [Accepted: 04/11/2012] [Indexed: 11/08/2022]
Abstract
Human immunodeficiency virus (HIV) protease inhibitors (PIs) produce profound and unpredictable drug-drug interactions (DDIs) that cannot be explained fully by their inhibition/inactivation of CYP3A enzymes. Delineating and quantifying the CYPs and transporters inducible by PIs are crucial in developing an integrative mechanistic understanding and prediction of PI-based DDIs. To do so, two LC-MS/MS cocktail assays were modified and validated simultaneously to quantify the CYP activity of CYP3A, 2B6, 2C8, 2C9, 2C19, 1A, 2E1, 2A6 and 2D6 enzymes. These new assays were applied to evaluate the induction potential of eight PIs in microsomes isolated from PI-treated human hepatocytes. The mRNA expression of these CYPs and transporters (OATP1B1, OATP1B3, OATP1A2, MDR1, MRP2 and MRP4) was also evaluated using relative RT-PCR. The majority of PIs were net inducers of CYP3As and 2B6 at both the mRNA and activity level (> 2-fold), while ritonavir, saquinavir, nelfinavir or lopinavir did not induce CYP3A activity (< 2-fold), presumably due to CYP3A inactivation. OATP1B1 and MDR1 were the only two hepatic transporters induced (> 2-fold) by the PIs. Amprenavir was the most potent net inducer. In conclusion, our validated cocktail assays can be implemented to comprehensively quantify CYP activities in human liver microsomes and hepatocyte studies. The results also provide the much needed data on the net induction potential of the PIs for hepatic CYPs and transporters. A qualitative agreement was observed between our results and published PI-based DDIs, suggesting that human hepatocytes are a useful platform for more extensive and quantitative in vitro-in vivo prediction of PI-based DDIs.
Collapse
Affiliation(s)
- Li Liu
- Department of Pharmaceutics, University of Washington, Seattle, 98195, USA
| | | | | | | | | | | |
Collapse
|
88
|
Kirby BJ, Collier AC, Kharasch ED, Whittington D, Thummel KE, Unadkat JD. Complex drug interactions of the HIV protease inhibitors 3: effect of simultaneous or staggered dosing of digoxin and ritonavir, nelfinavir, rifampin, or bupropion. Drug Metab Dispos 2012; 40:610-6. [PMID: 22190694 PMCID: PMC3286266 DOI: 10.1124/dmd.111.042705] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 12/05/2011] [Indexed: 01/27/2023] Open
Abstract
As part of a larger clinical drug-drug interaction (DDI) study aimed at in vitro to in vivo prediction of HIV protease inhibitor metabolic and transporter-based DDIs, we measured the inductive (staggered administration) and inductive plus inhibitory (simultaneously administered) effect of multiple dose ritonavir (RTV), nelfinavir (NFV), or rifampin (RIF) on the pharmacokinetics of the P-glycoprotein probe, digoxin (DIG), when administered simultaneously or staggered with the protease inhibitors or RIF. In both cases, NFV did not significantly affect DIG disposition. RTV decreased DIG renal clearance (Cl(renal)) when administered simultaneously or staggered but significantly increased DIG area under the curve from time zero to 24 h (AUC(0-24 h)) only when administered simultaneously. RIF decreased DIG AUC(0-24 h) only when RIF and DIG administration was staggered. When RIF and DIG were administered simultaneously, DIG maximal observed plasma concentration and area under the curve from time zero to 4 h were significantly increased, and DIG Cl(renal) was decreased. An unexpected and potentially clinically significant DDI was observed between DIG and the CYP2B6 probe, bupropion, which decreased DIG AUC(0-24 h) 1.6-fold and increased Cl(renal) 1.8-fold. Because this was an unexpected DDI and our studies were not specifically designed to quantify this interaction, further studies are required to confirm the interaction and understand the mechanistic basis of the DDI. In summary, RTV or NFV do not induce P-glycoprotein activity measured with DIG, and RIF does so only under staggered administration.
Collapse
Affiliation(s)
- Brian J Kirby
- Department of Pharmaceutics, University of Washington, Seattle,Washington 98195, USA
| | | | | | | | | | | |
Collapse
|
89
|
Kharasch ED, Bedynek PS, Hoffer C, Walker A, Whittington D. Lack of indinavir effects on methadone disposition despite inhibition of hepatic and intestinal cytochrome P4503A (CYP3A). Anesthesiology 2012; 116:432-47. [PMID: 22273859 PMCID: PMC3586934 DOI: 10.1097/aln.0b013e3182423478] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Methadone disposition and pharmacodynamics are highly susceptible to interactions with antiretroviral drugs. Methadone clearance and drug interactions have been attributed to cytochrome P4503A4 (CYP3A4), but actual mechanisms are unknown. Drug interactions can be clinically and mechanistically informative. This investigation assessed effects of the protease inhibitor indinavir on methadone pharmacokinetics and pharmacodynamics, hepatic and intestinal CYP3A4/5 activity (using alfentanil), and intestinal transporter activity (using fexofenadine). METHODS Twelve healthy volunteers underwent a sequential crossover. On three consecutive days they received oral alfentanil plus fexofenadine, intravenous alfentanil, and intravenous plus oral (deuterium-labeled) methadone. This was repeated after 2 weeks of indinavir. Plasma and urine analytes were measured by mass spectrometry. Opioid effects were measured by miosis. RESULTS Indinavir significantly inhibited hepatic and first-pass CYP3A activity. Intravenous alfentanil systemic clearance and hepatic extraction were reduced to 40-50% of control, apparent oral clearance to 30% of control, and intestinal extraction decreased by half, indicating 50% and 70% inhibition of hepatic and first-pass CYP3A activity. Indinavir increased fexofenadine area under the plasma concentration-time curve 3-fold, suggesting significant P-glycoprotein inhibition. Indinavir had no significant effects on methadone plasma concentrations, methadone N-demethylation, systemic or apparent oral clearance, renal clearance, hepatic extraction or clearance, or bioavailability. Methadone plasma concentration-effect relationships were unaffected by indinavir. CONCLUSIONS Despite significant inhibition of hepatic and intestinal CYP3A activity, indinavir had no effect on methadone N-demethylation and clearance, suggesting little or no role for CYP3A in clinical disposition of single-dose methadone. Inhibition of gastrointestinal transporter activity had no influence of methadone bioavailability.
Collapse
Affiliation(s)
- Evan D Kharasch
- Division of Clinical and Translational Research, Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63110-1093, USA.
| | | | | | | | | |
Collapse
|
90
|
Kirby BJ, Collier AC, Kharasch ED, Dixit V, Desai P, Whittington D, Thummel KE, Unadkat JD. Complex drug interactions of HIV protease inhibitors 2: in vivo induction and in vitro to in vivo correlation of induction of cytochrome P450 1A2, 2B6, and 2C9 by ritonavir or nelfinavir. Drug Metab Dispos 2011; 39:2329-37. [PMID: 21930825 PMCID: PMC3226381 DOI: 10.1124/dmd.111.038646] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 09/06/2011] [Indexed: 11/22/2022] Open
Abstract
Drug-drug interactions (DDIs) with the HIV protease inhibitors (PIs) are complex, paradoxical (e.g., ritonavir/alprazolam), and involve multiple mechanisms. As part of a larger study to better understand these DDIs and to devise a framework for in vitro to in vivo prediction of these DDIs, we determined the inductive effect of ∼2 weeks of administration of two prototypic PIs, nelfinavir (NFV), ritonavir (RTV), and rifampin (RIF; induction positive control) on the cytochrome P450 enzymes CYP1A2, CYP2B6, CYP2C9, and CYP2D6 and the inductive or inductive plus inhibitory effect of NFV, RTV, or RIF on CYP3A and P-glycoprotein in healthy human volunteers. Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Moreover, we accurately predicted the in vivo induction of these enzymes by quantifying their induction by the PIs in human hepatocytes and by using RIF as an in vitro to in vivo scalar. On the basis of the modest in vivo induction of CYP1A2, CYP2B6, or CYP2C9, the in vivo paradoxical DDIs with the PIs are likely explained by mechanisms other than induction of these enzymes such as induction of other metabolic enzymes, transporters, or both.
Collapse
Affiliation(s)
- Brian J Kirby
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Chen Y, Liu L, Monshouwer M, Fretland AJ. Determination of time-dependent inactivation of CYP3A4 in cryopreserved human hepatocytes and assessment of human drug-drug interactions. Drug Metab Dispos 2011; 39:2085-92. [PMID: 21835977 DOI: 10.1124/dmd.111.040634] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Assessment of time-dependent inhibition (TDI), especially CYP3A4, is an important parameter for preclinical and clinical development. The use of human liver microsomes (HLM) is the most common in vitro matrix to assess TDI, but this often leads to an overprediction of an actual effect observed clinically. Recently, the use of human hepatocytes has been hypothesized as a more relevant and possibly predictive matrix for the assessment of CYP3A4 TDI. Our work evaluates and optimizes three different human hepatocyte assays for the assessment of CYP3A4 TDI using pooled cryopreserved human hepatocytes. Using two of the optimized methods, the time-dependent inhibition kinetic parameters (K(I) and k(inact)) for four known CYP3A4 TDI (diltiazem, erythromycin, verapamil, and troleandomycin) were determined. When comparing TDI in HLM, the K(I) values from hepatocytes were in general 4- to 13-fold higher than that in HLM, whereas the k(inact) values in human hepatocytes were similar or slightly higher or lower depending on the inhibitor. The inactivation potency (k(inact)/K(I)) for four tested CYP3A4 inactivators in human hepatocytes was generally lower than that in HLM due to either lower affinity (K(I)) or lower inactivation rate (k(inact)) or both. When drug interactions were simulated with Simcyp using either HLM or human hepatocyte data, the predictions using the kinetic parameters from human hepatocytes resulted in a much better simulated change in pharmacokinetics compared with observed clinical data.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Drug Metabolism and Pharmacokinetics, Roche-Palo Alto, LLC, California, USA
| | | | | | | |
Collapse
|
92
|
Templeton IE, Houston JB, Galetin A. Predictive utility of in vitro rifampin induction data generated in fresh and cryopreserved human hepatocytes, Fa2N-4, and HepaRG cells. Drug Metab Dispos 2011; 39:1921-9. [PMID: 21771933 DOI: 10.1124/dmd.111.040824] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Rifampin is a potent inducer of CYP3A4 in vitro and precipitates numerous drug-drug interactions (DDIs) when coadministered with CYP3A4 substrates. In the current study, we have critically assessed reported rifampin in vitro CYP3A4 induction data in Fa2N-4, HepaRG, and cryopreserved or primary human hepatocytes, using either CYP3A4 mRNA or probe substrate metabolism as induction endpoints. An in vivo data base of intravenously administered victim drugs (assuming hepatic induction only) was collated (n = 18) to assess the predictive utility of these in vitro systems and to optimize rifampin in vivo E(max). In addition, the effect of substrate hepatic extraction ratio on prediction accuracy was investigated using prediction boundaries proposed recently (Drug Metab Dispos 39:170-173). Incorporation of hepatic extraction ratio in the prediction model resulted in accurate prediction of 89% of intravenous induction DDIs (n = 18), regardless of the in vitro system or induction endpoint (mRNA or CYP3A4 activity). Effects of in vitro parameters from different cellular systems, and optimized in vivo E(max), on the prediction of 21 oral DDIs were assessed. Use of mRNA data resulted in pronounced overprediction across all systems, with 86 to 100% of DDIs outside the acceptable prediction limits; in contrast, CYP3A4 activity predicted up to 62% of the oral DDIs within limits. Although prediction accuracy of oral DDIs was improved when using intravenous optimized rifampin E(max), >35% of DDIs were incorrectly assigned, suggesting potential differential E(max) between intestine and liver. Implications of the findings and recommendations for prediction of rifampin DDIs are discussed.
Collapse
Affiliation(s)
- Ian E Templeton
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Manchester, UK
| | | | | |
Collapse
|