1
|
Huang H, Zhao W, Qin N, Duan X. Recent Progress on Physiologically Based Pharmacokinetic (PBPK) Model: A Review Based on Bibliometrics. TOXICS 2024; 12:433. [PMID: 38922113 PMCID: PMC11209072 DOI: 10.3390/toxics12060433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Physiologically based pharmacokinetic/toxicokinetic (PBPK/PBTK) models are designed to elucidate the mechanism of chemical compound action in organisms based on the physiological, biochemical, anatomical, and thermodynamic properties of organisms. After nearly a century of research and practice, good results have been achieved in the fields of medicine, environmental science, and ecology. However, there is currently a lack of a more systematic review of progress in the main research directions of PBPK models, especially a more comprehensive understanding of the application in aquatic environmental research. In this review, a total of 3974 articles related to PBPK models from 1996 to 24 March 2024 were collected. Then, the main research areas of the PBPK model were categorized based on the keyword co-occurrence maps and cluster maps obtained by CiteSpace. The results showed that research related to medicine is the main application area of PBPK. Four major research directions included in the medical field were "drug assessment", "cross-species prediction", "drug-drug interactions", and "pediatrics and pregnancy drug development", in which "drug assessment" accounted for 55% of the total publication volume. In addition, bibliometric analyses indicated a rapid growth trend in the application in the field of environmental research, especially in predicting the residual levels in organisms and revealing the relationship between internal and external exposure. Despite facing the limitation of insufficient species-specific parameters, the PBPK model is still an effective tool for improving the understanding of chemical-biological effectiveness and will provide a theoretical basis for accurately assessing potential risks to ecosystems and human health. The combination with the quantitative structure-activity relationship model, Bayesian method, and machine learning technology are potential solutions to the previous research gaps.
Collapse
Affiliation(s)
| | | | - Ning Qin
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; (H.H.); (W.Z.)
| | - Xiaoli Duan
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; (H.H.); (W.Z.)
| |
Collapse
|
2
|
Dai X, Karol MD, Hitron M, Hard ML, Goulet MT, McLaughlin CF, Brantley SJ. Napabucasin Drug-Drug Interaction Potential, Safety, Tolerability, and Pharmacokinetics Following Oral Dosing in Healthy Adult Volunteers. Clin Pharmacol Drug Dev 2021; 10:824-839. [PMID: 34107166 PMCID: PMC8453567 DOI: 10.1002/cpdd.961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 04/10/2021] [Indexed: 12/05/2022]
Abstract
Napabucasin is an orally administered reactive oxygen species generator that is bioactivated by the intracellular antioxidant nicotinamide adenine dinucleotide phosphate:quinone oxidoreductase 1. Napabucasin induces cell death in cancer cells, including cancer stem cells. This phase 1 study (NCT03411122) evaluated napabucasin drug‐drug interaction potential for 7 cytochrome P450 (CYP) enzymes and the breast cancer resistance protein transporter/organic anion transporter 3. Healthy volunteers who tolerated napabucasin during period 1 received probe drugs during period 2, and in period 3 received napabucasin (240 mg twice daily; days 1‐11) plus a phenotyping cocktail containing omeprazole (CYP2C19), caffeine (CYP1A2), flurbiprofen (CYP2C9), bupropion (CYP2B6), dextromethorphan (CYP2D6), midazolam (CYP3A) (all oral; day 6), intravenous midazolam (day 7), repaglinide (CYP2C8; day 8), and rosuvastatin (breast cancer resistance protein/organic anion transporter 3; day 9). Drug‐drug interaction potential was evaluated in 17 of 30 enrolled volunteers. Napabucasin coadministration increased the area under the plasma concentration–time curve from time 0 extrapolated to infinity (geometric mean ratio [90% confidence interval]) of caffeine (124% [109.0%‐141.4%]), intravenous midazolam (118% [94.4%‐147.3%]), repaglinide (127% [104.7%‐153.3%]), and rosuvastatin (213% [42.5%‐1068.3%]) and decreased the area under the plasma concentration–time curve from time 0 extrapolated to infinity of dextromethorphan (71% [47.1%‐108.3%]), bupropion (79% [64.6%‐97.0%]), and hydroxybupropion (45% [15.7%‐129.6%]). No serious adverse events/deaths were reported. Generally, napabucasin is not expected to induce/inhibit drug clearance to a clinically meaningful degree.
Collapse
Affiliation(s)
- Xiaoshu Dai
- Clinical Pharmacology and Drug Metabolism, Sumitomo Dainippon Pharma Oncology, Inc., Cambridge, Massachusetts, USA
| | - Michael D Karol
- Clinical Pharmacology and Drug Metabolism, Sumitomo Dainippon Pharma Oncology, Inc., Cambridge, Massachusetts, USA
| | - Matthew Hitron
- Clinical Development, Sumitomo Dainippon Pharma Oncology, Inc., Cambridge, Massachusetts, USA
| | - Marjie L Hard
- Clinical Pharmacology, Nuventra, Inc., Durham, North Carolina, USA.,Current address: Praxis Precision Medicines, Cambridge, Massachusetts, USA
| | - Matthew T Goulet
- Clinical Pharmacology, Nuventra, Inc., Durham, North Carolina, USA.,Current address: Program Management Department, KSQ Therapeutics, Cambridge, Massachusetts, USA
| | | | - Scott J Brantley
- Clinical NCA & PK/PD, Nuventra, Inc., Durham, North Carolina, USA
| |
Collapse
|
3
|
Xu Y, Li Z. Imaging metabotropic glutamate receptor system: Application of positron emission tomography technology in drug development. Med Res Rev 2019; 39:1892-1922. [DOI: 10.1002/med.21566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Youwen Xu
- Independent Consultant and Contractor, Radiopharmaceutical Development, Validation and Bio-Application; Philadelphia Pennsylvania
| | - Zizhong Li
- Pharmaceutical Research and Development, SOFIE Biosciences; Somerset New Jersey
| |
Collapse
|
4
|
Zhang F, Bartels M, Clark A, Erskine T, Auernhammer T, Bhhatarai B, Wilson D, Marty S. Performance evaluation of the GastroPlus TM software tool for prediction of the toxicokinetic parameters of chemicals. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2018; 29:875-893. [PMID: 30286617 DOI: 10.1080/1062936x.2018.1518928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/30/2018] [Indexed: 06/08/2023]
Abstract
The accurate prediction of toxicokinetic parameters arising from oral, dermal and inhalation routes of chemical exposure is a key element in chemical safety assessments. In this research, the physiologically based pharmacokinetic (PBPK) GastroPlusTM software was evaluated against a series of chemicals for the prediction of toxicokinetic parameters. Overall, 67% of predicted intrinsic clearance (Clint) values were within 1- to 10-fold of empirical data for 463 compounds, and 87% of the predicted fraction unbounded in plasma (Fup) values were 1- to 3-fold of empirical data for 441 compounds. The r2 (coefficient of determination) of predicted Cmax (maximum plasma concentration) and AUC (Area Under Curve) values versus the corresponding empirical values from oral, inhalation and dermal exposures ranged from 0.04 to 0.92. Among the three exposures, the highest r2 values, ranging from 0.80 to 0.92, were observed for oral exposure predictions, where 88% of the compounds had 1- to 10-fold differences between predicted and empirical values for Cmax and AUC. The predicted plasma Css (steady-state plasma concentration) values were consistent with those Css values calculated by in vitro-to-in vivo extrapolation (IVIVE) approaches using experimental parameters. Based on the evaluation results, GastroPlus™ can be used as a QSAR/PBPK tool for toxicokinetic parameter predictions.
Collapse
Affiliation(s)
- F Zhang
- a The Dow Chemical Company , Midland , MI , USA
| | - M Bartels
- b ToxMetrics.com LLC , Midland , MI , USA
| | - A Clark
- a The Dow Chemical Company , Midland , MI , USA
| | - T Erskine
- a The Dow Chemical Company , Midland , MI , USA
| | | | - B Bhhatarai
- c Novartis Institute for Biomedical Research , Cambridge , MA , USA
| | - D Wilson
- a The Dow Chemical Company , Midland , MI , USA
| | - S Marty
- a The Dow Chemical Company , Midland , MI , USA
| |
Collapse
|
5
|
Narushima K, Maeda H, Shiramoto M, Endo Y, Ohtsuka S, Nakamura H, Nagata Y, Uchimura T, Kannami A, Shimazaki R, Fukagawa M, Akizawa T. Assessment of CYP-Mediated Drug Interactions for Evocalcet, a New Calcimimetic Agent, Based on In Vitro Investigations and a Cocktail Study in Humans. Clin Transl Sci 2018; 12:20-27. [PMID: 30238620 PMCID: PMC6342237 DOI: 10.1111/cts.12588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/21/2018] [Indexed: 01/25/2023] Open
Abstract
Evocalcet is a novel calcimimetic agent for the treatment of secondary hyperparathyroidism (SHPT). This study evaluated the effects of evocalcet on inhibition and induction of cytochrome P450 (CYP) isozymes. Although drug interactions arising from reversible inhibition of CYP isozymes by evocalcet were considered unlikely based on the results of in vitro studies and static model analyses, the potential for evocalcet to cause time‐dependent inhibition of CYP3A or induction of several CYP isozymes could not be ruled out. Therefore, a clinical drug‐drug interaction (DDI) study to evaluate the effects of evocalcet on the pharmacokinetics (PKs) of probe substrates for CYP isozymes (CYP1A2, CYP2B6, CYP2C8, CYP2C9, and CYP3A) was conducted in healthy male volunteers using a novel cocktail combination. Evocalcet did not significantly affect the PKs of the probe substrates, confirming that CYP‐mediated interactions were unlikely.
Collapse
Affiliation(s)
| | - Hiroshi Maeda
- R&D Division, Kyowa Hakko Kirin Co. Ltd., Tokyo, Japan
| | | | - Yuichi Endo
- R&D Division, Kyowa Hakko Kirin Co. Ltd., Tokyo, Japan
| | | | | | | | | | - Ayako Kannami
- DMPK Research Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | | | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Tadao Akizawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Perkins EJ, Posada M, Kellie Turner P, Chappell J, Ng WT, Twelves C. Physiologically Based Pharmacokinetic Modelling of Cytochrome P450 2C9-Related Tolbutamide Drug Interactions with Sulfaphenazole and Tasisulam. Eur J Drug Metab Pharmacokinet 2018; 43:355-367. [PMID: 29119333 PMCID: PMC5956062 DOI: 10.1007/s13318-017-0447-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Background and Objectives Cytochrome P450 2C9 (CYP2C9) is involved in the biotransformation of many commonly used drugs, and significant drug interactions have been reported for CYP2C9 substrates. Previously published physiologically based pharmacokinetic (PBPK) models of tolbutamide are based on an assumption that its metabolic clearance is exclusively through CYP2C9; however, many studies indicate that CYP2C9 metabolism is only responsible for 80–90% of the total clearance. Therefore, these models are not useful for predicting the magnitude of CYP2C9 drug–drug interactions (DDIs). This paper describes the development and verification of SimCYP®-based PBPK models that accurately describe the human pharmacokinetics of tolbutamide when dosed alone or in combination with the CYP2C9 inhibitors sulfaphenazole and tasisulam. Methods A PBPK model was optimized in SimCYP® for tolbutamide as a CYP2C9 substrate, based on published in vitro and clinical data. This model was verified to replicate the magnitude of DDI reported with sulfaphenazole and was further applied to simulate the DDI with tasisulam, a small molecule investigated for the treatment of cancer. A clinical study (CT registration # NCT01185548) was conducted in patients with cancer to assess the pharmacokinetic interaction of tasisulum with tolbutamide. A PBPK model was built for tasisulam, and the clinical study design was replicated using the optimized tolbutamide model. Results The optimized tolbutamide model accurately predicted the magnitude of tolbutamide AUC increase (5.3–6.2-fold) reported for sulfaphenazole. Furthermore, the PBPK simulations in a healthy volunteer population adequately predicted the increase in plasma exposure of tolbutamide in patients with cancer (predicted AUC ratio = 4.7–5.4; measured mean AUC ratio = 5.7). Conclusions This optimized tolbutamide PBPK model was verified with two strong CYP2C9 inhibitors and can be applied to the prediction of CYP2C9 interactions for novel inhibitors. Furthermore, this work highlights the utility of mechanistic models in navigating the challenges in conducting clinical pharmacology studies in cancer patients.
Collapse
|
7
|
Fuhr U, Hsin CH, Li X, Jabrane W, Sörgel F. Assessment of Pharmacokinetic Drug-Drug Interactions in Humans: In Vivo Probe Substrates for Drug Metabolism and Drug Transport Revisited. Annu Rev Pharmacol Toxicol 2018; 59:507-536. [PMID: 30156973 DOI: 10.1146/annurev-pharmtox-010818-021909] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pharmacokinetic parameters of selective probe substrates are used to quantify the activity of an individual pharmacokinetic process (PKP) and the effect of perpetrator drugs thereon in clinical drug-drug interaction (DDI) studies. For instance, oral caffeine is used to quantify hepatic CYP1A2 activity, and oral dagibatran etexilate for intestinal P-glycoprotein (P-gp) activity. However, no probe substrate depends exclusively on the PKP it is meant to quantify. Lack of selectivity for a given enzyme/transporter and expression of the respective enzyme/transporter at several sites in the human body are the main challenges. Thus, a detailed understanding of the role of individual PKPs for the pharmacokinetics of any probe substrate is essential to allocate the effect of a perpetrator drug to a specific PKP; this is a prerequisite for reliably informed pharmacokinetic models that will allow for the quantitative prediction of perpetrator effects on therapeutic drugs, also in respective patient populations not included in DDI studies.
Collapse
Affiliation(s)
- Uwe Fuhr
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Chih-Hsuan Hsin
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Xia Li
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Wafaâ Jabrane
- Department I of Pharmacology, University Hospital Cologne, 50931 Cologne, Germany;
| | - Fritz Sörgel
- Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany
| |
Collapse
|
8
|
Design and optimization of the cocktail assay for rapid assessment of the activity of UGT enzymes in human and rat liver microsomes. Toxicol Lett 2018; 295:379-389. [PMID: 30036684 DOI: 10.1016/j.toxlet.2018.07.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/21/2018] [Accepted: 07/19/2018] [Indexed: 12/26/2022]
Abstract
Along with the prevalence of drug combination therapies, an increasing number of cases about drug-drug interactions (DDI) have been reported, which has drawn a lot of attention due to the potential toxicity and/or therapeutic failure. Pharmacokinetic interactions based on drug metabolic enzymes should be responsible for a great many of DDI. UDP-glucuronosyltransferases (UGT) as the main phase II metabolic enzymes are involved in the metabolism of many endogenous and exogenous substrates. Herein, we designed and optimized a validated cocktail method for the simultaneous evaluation of drug-mediated inhibition of the main five UGT isoforms using respective specific probe substrates (estradiol for UGT1A1, chenodeoxycholic acid for UGT1A3, serotonin for UGT1A6, propofol for UGT1A9/PROG and zidovudine for UGT2B7/AZTG) in human and rat liver microsomes by liquid chromatography-tandem mass spectrometry (LCMS/MS). Moreover, we investigated the risk of interactions among UGT probe substrates, and validated the cocktail method by known positive inhibitors of UGT isoforms. To minimize the substrates interaction, we developed two cocktail subgroups which were further optimized via exploring the experimental conditions. In particular, the cocktail inhibition assay for rapid assessment of in vitro rat UGTs was firstly reported and the values of Km in the liver microsomes from humans and rats were close to each other in the specific UGT subtype. In conclusion, this study has successfully established the cocktail approach to explore UGT activity, especially for UGT inhibition in a fast and efficient way.
Collapse
|
9
|
Prediction of drug–drug interaction potential using physiologically based pharmacokinetic modeling. Arch Pharm Res 2017; 40:1356-1379. [DOI: 10.1007/s12272-017-0976-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022]
|
10
|
Budha NR, Ji T, Musib L, Eppler S, Dresser M, Chen Y, Jin JY. Evaluation of Cytochrome P450 3A4-Mediated Drug-Drug Interaction Potential for Cobimetinib Using Physiologically Based Pharmacokinetic Modeling and Simulation. Clin Pharmacokinet 2017; 55:1435-1445. [PMID: 27225997 DOI: 10.1007/s40262-016-0412-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVES Cobimetinib is eliminated mainly through cytochrome P450 (CYP) 3A4-mediated hepatic metabolism in humans. A clinical drug-drug interaction (DDI) study with the potent CYP3A4 inhibitor itraconazole resulted in an approximately sevenfold increase in cobimetinib exposure. The DDI risk for cobimetinib with other CYP3A4 inhibitors and inducers needs to be assessed in order to provide dosing instructions. METHODS A physiologically based pharmacokinetic (PBPK) model was developed for cobimetinib using in vitro data. It was then optimized and verified using clinical pharmacokinetic data and itraconazole-cobimetinib DDI data. The contribution of CYP3A4 to the clearance of cobimetinib in humans was confirmed using sensitivity analysis in a retrospective simulation of itraconazole-cobimetinib DDI data. The verified PBPK model was then used to predict the effect of other CYP3A4 inhibitors and inducers on cobimetinib pharmacokinetics. RESULTS The PBPK model described cobimetinib pharmacokinetic profiles after both intravenous and oral administration of cobimetinib well and accurately simulated the itraconazole-cobimetinib DDI. Sensitivity analysis suggested that CYP3A4 contributes ~78 % of the total clearance of cobimetinib. The PBPK model predicted no change in cobimetinib exposure (area under the plasma concentration-time curve, AUC) with the weak CYP3A inhibitor fluvoxamine and a three to fourfold increase with the moderate CYP3A inhibitors, erythromycin and diltiazem. Similarly, cobimetinib exposure in the presence of strong (rifampicin) and moderate (efavirenz) CYP3A inducers was predicted to decrease by 83 and 72 %, respectively. CONCLUSION This study demonstrates the value of using PBPK simulation to assess the clinical DDI risk inorder to provide dosing instructions with other CYP3A4 perpetrators.
Collapse
Affiliation(s)
- Nageshwar R Budha
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Tao Ji
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Luna Musib
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Steve Eppler
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Mark Dresser
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| | - Yuan Chen
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Jin Y Jin
- Clinical Pharmacology, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
11
|
Rabinovich-Guilatt L, Steiner L, Hallak H, Pastino G, Muglia P, Spiegelstein O. Metoprolol-pridopidine drug-drug interaction and food effect assessments of pridopidine, a new drug for treatment of Huntington's disease. Br J Clin Pharmacol 2017; 83:2214-2224. [PMID: 28449367 PMCID: PMC5595947 DOI: 10.1111/bcp.13317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/24/2017] [Accepted: 04/18/2017] [Indexed: 01/22/2023] Open
Abstract
Aims Pridopidine is an oral drug in clinical development for treatment of patients with Huntington's disease. This study examined the interactions of pridopidine with in vitro cytochrome P450 activity and characterized the effects of pridopidine on CYP2D6 activity in healthy volunteers using metoprolol as a probe substrate. The effect of food on pridopidine exposure was assessed. Methods The ability of pridopidine to inhibit and/or induce in vitro activity of drug metabolizing enzymes was examined in human liver microsomes and fresh hepatocytes. CYP2D6 inhibition potency and reversibility was assessed using dextromethorphan. For the clinical assessment, 22 healthy subjects were given metoprolol 100 mg alone and concomitantly with steady‐state pridopidine 45 mg twice daily. Food effect on a single 90 mg dose of pridopidine was evaluated in a crossover manner. Safety assessments and pharmacokinetic sampling occurred throughout the study. Results Pridopidine was found to be a metabolism dependent inhibitor of CYP2D6, the main enzyme catalysing its own metabolism. Flavin‐containing monooxygenase heat inactivation of liver microsomes did not affect pridopidine metabolism‐dependent inhibition of CYP2D6 and its inhibition of CYP2D6 was not reversible with addition of FeCN3. Exposure to metoprolol was markedly increased when coadministered with pridopidine; the ratio of the geometric means (90% confidence interval) for maximum observed plasma concentration, and area under the plasma concentration–time curve from time 0 to the time of the last quantifiable concentration and extrapolated to infinity were 3.5 (2.9, 4.22), 6.64 (5.27, 8.38) and 6.55 (5.18, 8.28), respectively. Systemic exposure to pridopidine was unaffected by food conditions. Conclusions As pridopidine is a metabolism‐dependent inhibitor of CYP2D6, systemic levels of drugs metabolized by CYP2D6 may increase with chronic coadministration of pridopidine. Pridopidine can be administered without regard to food.
Collapse
Affiliation(s)
| | - Lilach Steiner
- Drug Metabolism and Pharmacokinetics, Teva Pharmaceutical Industries Ltd, Netanya, Israel
| | - Hussein Hallak
- Drug Metabolism and Pharmacokinetics, Teva Pharmaceutical Industries Ltd, Netanya, Israel
| | - Gina Pastino
- Clinical Pharmacology & Pharmacometrics, Teva Pharmaceutical Industries Ltd, Malvern PA, USA
| | - Pierandrea Muglia
- Neuroscience Discovery Medicine UCB Pharma Chemin du Foriest, Belgium
| | - Ofer Spiegelstein
- Clinical Pharmacology & Pharmacometrics, Teva Pharmaceutical Industries Ltd, Netanya, Israel
| |
Collapse
|
12
|
Liu L, Miao MX, Zhong ZY, Xu P, Chen Y, Liu XD. Chronic administration of caderofloxacin, a new fluoroquinolone, increases hepatic CYP2E1 expression and activity in rats. Acta Pharmacol Sin 2016; 37:561-70. [PMID: 26838075 DOI: 10.1038/aps.2015.160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
AIM Caderofloxacin is a new fluoroquinolone that is under phase III clinical trials in China. Here we examined the effects of caderofloxacin on rat hepatic cytochrome P450 (CYP450) isoforms as well as the potential of caderofloxacin interacting with co-administered drugs. METHODS Male rats were treated with caderofloxacin (9 mg/kg, ig) once or twice daily for 14 consecutive days. The effects of caderofloxacin on CYP3A, 2D6, 2C19, 1A2, 2E1 and 2C9 were evaluated using a "cocktail" of 6 probes (midazolam, dextromethorphan, omeprazole, theophylline, chlorzoxazone and diclofenac) injected on d 0 (prior to caderofloxacin exposure) and d 15 (after caderofloxacin exposure). Hepatic microsomes from the caderofloxacin-treated rats were used to assess CYP2E1 activity and chlorzoxazone metabolism. The expression of CYP2E1 mRNA and protein in hepatic microsomes was analyzed with RT-PCR and Western blotting, respectively. RESULTS Fourteen-day administration of caderofloxacin significantly increased the activity of hepatic CYP2E1, leading to enhanced metabolism of chlorzoxazone. In vitro microsomal study confirmed that CYP2E1 was a major metabolic enzyme involved in chlorzoxazone metabolism, and the 14-d administration of caderofloxacin significantly increased the activity of CYP2E1 in hepatic microsomes, resulting in increased formation of 6-hydroxychlorzoxazone. Furthermore, the 14-d administration of caderofloxacin significantly increased the expression of CYP2E1 mRNA and protein in liver microsomes, which was consistent with the pharmacokinetic results. CONCLUSION Fourteen-day administration of caderofloxacin can induce the expression and activity of hepatic CYP2E1 in rats. When caderofloxacin is administered, a potential drug-drug interaction mediated by CYP2E1 induction should be considered.
Collapse
|
13
|
Wilk-Zasadna I, Bernasconi C, Pelkonen O, Coecke S. Biotransformation in vitro: An essential consideration in the quantitative in vitro-to-in vivo extrapolation (QIVIVE) of toxicity data. Toxicology 2014; 332:8-19. [PMID: 25456264 DOI: 10.1016/j.tox.2014.10.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 06/11/2014] [Accepted: 10/11/2014] [Indexed: 12/14/2022]
Abstract
Early consideration of the multiplicity of factors that govern the biological fate of foreign compounds in living systems is a necessary prerequisite for the quantitative in vitro-in vivo extrapolation (QIVIVE) of toxicity data. Substantial technological advances in in vitro methodologies have facilitated the study of in vitro metabolism and the further use of such data for in vivo prediction. However, extrapolation to in vivo with a comfortable degree of confidence, requires continuous progress in the field to address challenges such as e.g., in vitro evaluation of chemical-chemical interactions, accounting for individual variability but also analytical challenges for ensuring sensitive measurement technologies. This paper discusses the current status of in vitro metabolism studies for QIVIVE extrapolation, serving today's hazard and risk assessment needs. A short overview of the methodologies for in vitro metabolism studies is given. Furthermore, recommendations for priority research and other activities are provided to ensure further widespread uptake of in vitro metabolism methods in 21st century toxicology. The need for more streamlined and explicitly described integrated approaches to reflect the physiology and the related dynamic and kinetic processes of the human body is highlighted i.e., using in vitro data in combination with in silico approaches.
Collapse
Affiliation(s)
- Iwona Wilk-Zasadna
- Systems Toxicology Unit/EURL ECVAM, Institute for Health and Consumer Protection, European Commission Joint Research Centre, Ispra, Varese I-21027, Italy
| | - Camilla Bernasconi
- Systems Toxicology Unit/EURL ECVAM, Institute for Health and Consumer Protection, European Commission Joint Research Centre, Ispra, Varese I-21027, Italy
| | - Olavi Pelkonen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Sandra Coecke
- Systems Toxicology Unit/EURL ECVAM, Institute for Health and Consumer Protection, European Commission Joint Research Centre, Ispra, Varese I-21027, Italy.
| |
Collapse
|
14
|
Zang M, Zhu F, Li X, Yang A, Xing J. Auto-induction of phase I and phase II metabolism of artemisinin in healthy Chinese subjects after oral administration of a new artemisinin-piperaquine fixed combination. Malar J 2014; 13:214. [PMID: 24889062 PMCID: PMC4055232 DOI: 10.1186/1475-2875-13-214] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/27/2014] [Indexed: 11/25/2022] Open
Abstract
Background Artequick is a relatively inexpensive artemisinin (Qing-hao-su; QHS)-based combination therapy (ACT) that contains QHS and piperaquine (PQ), which has not been widely used because of the decreased concentration level of QHS after repeated oral administrations for five to seven days as a monotherapy. This study was designed to evaluate the potential auto-induction metabolism of QHS in healthy Chinese adults after a two-day oral administration of QHS-PQ. The effect of QHS-PQ on the activity of the CYP2B6 and CYP3A4 was also investigated. Methods Fourteen healthy Chinese subjects received two-day oral doses of QHS-PQ (Artequick). A two-drug cocktail consisting of bupropion and midazolam was used to assess the activities of CYP2B6 and CYP3A, respectively. Plasma samples were analysed for QHS and its phase I/II metabolites, probe drugs and their metabolites, using a validated liquid chromatography tandem mass spectrometric (LC-MS) method. Results Four major phase I metabolites of QHS (M1-M3 and deoxy-QHS) and two subsequent phase II metabolites (M4-M5) were detected in human plasma after oral administrations of QHS-PQ. The AUC0-t of the QHS and its phase I metabolites decreased significantly (P < 0.05) with increased oral clearance (CL/F) after two-day oral doses of QHS-PQ, whereas its phase II metabolites exhibited higher AUC (P < 0.01). The phase I metabolic capability, calculated by the AUC0-t ratio of all phase I metabolites to QHS, increased 1.5-fold after the repeated dose (P < 0.01), and the phase II metabolic capability increased 1.5-fold for M4 and 3.0-fold for M5. The enzyme activity of CYP2B6 and CYP3A4 increased 2.1-fold and 3.2-fold, respectively, after two-day oral doses of QHS-PQ. Conclusions The auto-induction of both phase I and phase II metabolism of QHS was present in healthy Chinese subjects after a recommended two-day oral dose of QHS-PQ. The auto-induction metabolism also existed for phase I metabolites of QHS. The enzyme activity of CYP2B6 and CYP3A4 was induced after the two-day oral doses of QHS-PQ. Based on these results, the alternative common three-day regimen for QHS-PQ could probably lead to lower bioavailability of QHS and higher potential of drug-drug interaction caused by the induction of drug-metabolizing enzymes.
Collapse
Affiliation(s)
| | | | | | | | - Jie Xing
- School of Pharmaceutical Sciences, Shandong University, 44# West Wenhua Road, Jinan 250012, P R China.
| |
Collapse
|