1
|
Lenard A, Hermann SA, Stoll F, Burhenne J, Foerster KI, Czock D, Mikus G, Meid AD, Haefeli WE, Blank A. Effect of the frequently used antiepileptic drugs carbamazepine, gabapentin, and pregabalin on the pharmacokinetics of edoxaban and other oral factor xa inhibitors in healthy volunteers. Front Pharmacol 2025; 16:1542063. [PMID: 40290437 PMCID: PMC12022901 DOI: 10.3389/fphar.2025.1542063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose Pregabalin, gabapentin, and carbamazepine, a potent inducer of cytochrome P450 (CYP) 3A4 and P-glycoprotein, are frequently used antiepileptic drugs that are often administered together with factor Xa inhibitors (FXaI). We aimed to investigate whether potentially clinically relevant drug-drug interactions occur with these combinations. Methods In an open-label fixed-sequence trial in 36 healthy volunteers, we evaluated the pharmacokinetics of 60 mg edoxaban and of a microdosed FXaI cocktail (25 µg apixaban, 50 µg edoxaban, and 25 µg rivaroxaban) before and during treatment with carbamazepine (12 evaluable volunteers, individually dosed to therapeutic concentrations), gabapentin (11 volunteers, titrated to 3 × 400 mg/d), and pregabalin (12 volunteers, titrated to 2 × 300 mg/d). The antiepileptics were dosed to steady-state and the CYP3A activity was evaluated by assessing the pharmacokinetics of microdosed midazolam (30 µg). Results Carbamazepine reduced the area under the plasma concentration-time curve (AUC ∞ ) of 60 mg edoxaban by a factor of 0.48 (geometric mean ratio (GMR) with 90% CI (0.41-0.56); p < 0.0001) and Cmax by a factor of 0.47 (0.34-0.66) and reduced the exposure of the edoxaban metabolite M-4 to a similar extent. Carbamazepine also decreased the exposure (AUC ∞ ) of microdosed apixaban, edoxaban, and rivaroxaban by a factor of 0.66, 0.59, and 0.56, respectively. Gabapentin and pregabalin did neither affect the exposure of 60 mg edoxaban nor the exposure of any microdosed FXaI. Conclusion Carbamazepine decreased FXaI exposure to a clinically relevant extent and dose adjustment may be required to maintain an adequate anticoagulant effect, whereas gabapentin and pregabalin do not require dose adjustment of FXaI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Antje Blank
- Internal Medicine IX, Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University, Medical Faculty Heidelberg/Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
2
|
Madrid MF, Mendoza EN, Padilla AL, Choquenaira-Quispe C, de Jesus Guimarães C, de Melo Pereira JV, Barros-Nepomuceno FWA, Lopes Dos Santos I, Pessoa C, de Moraes Filho MO, Rocha DD, Ferreira PMP. In vitro models to evaluate multidrug resistance in cancer cells: Biochemical and morphological techniques and pharmacological strategies. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2025; 28:1-27. [PMID: 39363148 DOI: 10.1080/10937404.2024.2407452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The overexpression of ATP-binding cassette (ABC) transporters contributes to the failure of chemotherapies and symbolizes a great challenge in oncology, associated with the adaptation of tumor cells to anticancer drugs such that these transporters become less effective, a mechanism known as multidrug resistance (MDR). The aim of this review is to present the most widely used methodologies for induction and comprehension of in vitro models for detection of multidrug-resistant (MDR) modulators or inhibitors, including biochemical and morphological techniques for chemosensitivity studies. The overexpression of MDR proteins, predominantly, the subfamily glycoprotein-1 (P-gp or ABCB1) multidrug resistance, multidrug resistance-associated protein 1 (MRP1 or ABCCC1), multidrug resistance-associated protein 2 (MRP2 or ABCC2) and cancer resistance protein (ABCG2), in chemotherapy-exposed cancer lines have been established/investigated by several techniques. Amongst these techniques, the most used are (i) colorimetric/fluorescent indirect bioassays, (ii) rhodamine and efflux analysis, (iii) release of 3,30-diethyloxacarbocyanine iodide by fluorescence microscopy and flow cytometry to measure P-gp function and other ABC transporters, (iv) exclusion of calcein-acetoxymethylester, (v) ATPase assays to distinguish types of interaction with ABC transporters, (vi) morphology to detail phenotypic characteristics in transformed cells, (vii) molecular testing of resistance-related proteins (RT-qPCR) and (viii) 2D and 3D models, (ix) organoids, and (x) microfluidic technology. Then, in vitro models for detecting chemotherapy MDR cells to assess innovative therapies to modulate or inhibit tumor cell growth and overcome clinical resistance. It is noteworthy that different therapies including anti-miRNAs, antibody-drug conjugates (to natural products), and epigenetic modifications were also considered as promising alternatives, since currently no anti-MDR therapies are able to improve patient quality of life. Therefore, there is also urgency for new clinical markers of resistance to more reliably reflect in vivo effectiveness of novel antitumor drugs.
Collapse
Affiliation(s)
- Maria Fernanda Madrid
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Eleicy Nathaly Mendoza
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Ana Lizeth Padilla
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
| | - Celia Choquenaira-Quispe
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
- Catholic University of Santa María, Arequipa, Perú
| | - Celina de Jesus Guimarães
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - João Victor de Melo Pereira
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Ingredy Lopes Dos Santos
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - Claudia Pessoa
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Manoel Odorico de Moraes Filho
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Danilo Damasceno Rocha
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
3
|
Yang Z, Qu Y, Sun Y, Pan J, Zhou T, Yu Y. Evaluation of Drug-Drug Interactions Between Clarithromycin and Direct Oral Anticoagulants Using Physiologically Based Pharmacokinetic Models. Pharmaceutics 2024; 16:1449. [PMID: 39598572 PMCID: PMC11597346 DOI: 10.3390/pharmaceutics16111449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Objective: This study assessed the pharmacokinetic (PK) interactions between clarithromycin (a P-glycoprotein [P-gp] inhibitor) and four direct oral anticoagulants (DOACs) (P-gp substrates) using physiologically based PK (PBPK) models to elucidate the influence of P-gp in the interaction between them. Methods: PBPK models for clarithromycin, DABE-dabigatran (DAB), rivaroxaban, apixaban, and edoxaban were constructed using GastroPlus™ (version 9.9), based on physicochemical data and PK parameters from the literature. The models were optimized and validated in healthy subjects. We evaluated the predictive performance of the established model and further assessed the impact of P-gp on the PK of the four DOACs. Successfully validated models were then used to evaluate potential drug-drug interactions (DDIs) between clarithromycin and the DOACs. Results: The established PBPK models accurately described the PK of clarithromycin, DABE-DAB, rivaroxaban, apixaban, and edoxaban. The predicted PK parameters (Cmax, Tmax, AUC0-t) were within 0.5-2 times the observed values. A sensitivity analysis of P-gp parameters indicated that an increase in P-gp expression was reduced by in vivo exposure to DOACs. The models demonstrated good predictive ability for DDIs between clarithromycin and the anticoagulants, and the ratio of the predicted values to the observed values of Cmax and the area under the curve (AUC) in the DDI state was within the range of 0.5-2. Conclusions: Comprehensive PBPK models for clarithromycin, DABE-DAB, rivaroxaban, apixaban, and edoxaban were developed, which can effectively predict DDIs mediated by P-gp's function. These models provide theoretical support for clinical dose adjustments and serve as a foundation for future PBPK model development for DOACs under specific pathological conditions.
Collapse
Affiliation(s)
- Zhuan Yang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; (Z.Y.); (Y.S.); (J.P.)
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Yuchen Qu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China;
| | - Yewen Sun
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; (Z.Y.); (Y.S.); (J.P.)
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; (Z.Y.); (Y.S.); (J.P.)
| | - Tong Zhou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Yunli Yu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; (Z.Y.); (Y.S.); (J.P.)
| |
Collapse
|
4
|
Șorodoc V, Indrei L, Dobroghii C, Asaftei A, Ceasovschih A, Constantin M, Lionte C, Morărașu BC, Diaconu AD, Șorodoc L. Amiodarone Therapy: Updated Practical Insights. J Clin Med 2024; 13:6094. [PMID: 39458044 PMCID: PMC11508869 DOI: 10.3390/jcm13206094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Amiodarone, a bi-iodinated benzofuran derivative, is among the most commonly used antiarrhythmic drugs due to its high level of effectiveness. Though initially categorized as a class III agent, amiodarone exhibits antiarrhythmic properties across all four classes of antiarrhythmic drugs. Amiodarone is highly effective in maintaining sinus rhythm in patients with paroxysmal atrial fibrillation while also playing a crucial role in preventing a range of ventricular arrhythmias. Amiodarone has a complex pharmacokinetic profile, characterized by a large volume of distribution and a long half-life, which can range from several weeks to months, resulting in prolonged effects even after discontinuation. Side effects may include thyroid dysfunction, pulmonary fibrosis, and hepatic injury, necessitating regular follow-ups. Additionally, amiodarone interacts with several drugs, including anticoagulants, which must be managed to prevent adverse effects. Therefore, a deep understanding of both oral and intravenous formulations, as well as proper dosage adjustments, is essential. The aim of this paper is to provide a comprehensive and updated review on amiodarone's indications, contraindications, recommended dosages, drug interactions, side effects, and monitoring protocols.
Collapse
Affiliation(s)
- Victorița Șorodoc
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Lucia Indrei
- Radiology and Medical Imaging Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Catinca Dobroghii
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Andreea Asaftei
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Alexandr Ceasovschih
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mihai Constantin
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Cătălina Lionte
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Bianca Codrina Morărașu
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Alexandra-Diana Diaconu
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Laurențiu Șorodoc
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Second Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
| |
Collapse
|
5
|
Lenard A, Hermann SA, Stoll F, Burhenne J, Foerster KI, Mikus G, Meid AD, Haefeli WE, Blank A. Effect of Clarithromycin, a Strong CYP3A and P-glycoprotein Inhibitor, on the Pharmacokinetics of Edoxaban in Healthy Volunteers and the Evaluation of the Drug Interaction with Other Oral Factor Xa Inhibitors by a Microdose Cocktail Approach. Cardiovasc Drugs Ther 2024; 38:747-756. [PMID: 36870039 PMCID: PMC11266212 DOI: 10.1007/s10557-023-07443-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 03/05/2023]
Abstract
PURPOSE We assessed the differential effect of clarithromycin, a strong inhibitor of cytochrome P450 (CYP) 3A4 and P-glycoprotein, on the pharmacokinetics of a regular dose of edoxaban and on a microdose cocktail of factor Xa inhibitors (FXaI). Concurrently, CYP3A activity was determined with a midazolam microdose. METHODS In an open-label fixed-sequence trial in 12 healthy volunteers, the pharmacokinetics of a microdosed FXaI cocktail (μ-FXaI; 25 μg apixaban, 50 μg edoxaban, and 25 μg rivaroxaban) and of 60 mg edoxaban before and during clarithromycin (2 x 500 mg/d) dosed to steady-state was evaluated. Plasma concentrations of study drugs were quantified using validated ultra-performance liquid chromatography-tandem mass spectrometry methods. RESULTS Therapeutic clarithromycin doses increased the exposure of a therapeutic 60 mg dose of edoxaban with a geometric mean ratio (GMR) of the area under the plasma concentration-time curve (AUC) of 1.53 (90 % CI: 1.37-1.70; p < 0.0001). Clarithromycin also increased the GMR (90% CI) of the exposure of microdosed FXaI apixaban to 1.38 (1.26-1.51), edoxaban to 2.03 (1.84-2.24), and rivaroxaban to 1.44 (1.27-1.63). AUC changes observed for the therapeutic edoxaban dose were significantly smaller than those observed with the microdose (p < 0.001). CONCLUSION Clarithromycin increases FXaI exposure. However, the magnitude of this drug interaction is not expected to be clinically relevant. The edoxaban microdose overestimates the extent of the drug interaction with the therapeutic dose, whereas AUC ratios for apixaban and rivaroxaban were comparable to the interaction with therapeutic doses as reported in the literature. TRIAL REGISTRATION EudraCT Number: 2018-002490-22.
Collapse
Affiliation(s)
- Alexander Lenard
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Simon A Hermann
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Felicitas Stoll
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Juergen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Kathrin I Foerster
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Andreas D Meid
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany
| | - Antje Blank
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- Partner Site Heidelberg, German Center for Infection Research, Heidelberg, Germany.
| |
Collapse
|
6
|
Chanteux H, MacPherson M, Kramer H, Otoul C, Okagaki T, Rospo C, De Bruyn S, Watling M, Bani M, Sciberras D. Overview of preclinical and clinical studies investigating pharmacokinetics and drug-drug interactions of padsevonil. Expert Opin Drug Metab Toxicol 2024; 20:841-855. [PMID: 38932723 DOI: 10.1080/17425255.2024.2373108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Padsevonil is an antiseizure medication candidate intended to benefit patients with drug-resistant epilepsy. Our investigations aimed at characterizing pharmacokinetics and drug-drug interaction (DDI) profile of padsevonil. RESEARCH DESIGN AND METHODS An overview of preclinical and clinical pharmacology studies conducted during padsevonil development is provided. RESULTS In preclinical studies, cytochrome (CYP) 3A4 was identified as the main P450 isoform involved in padsevonil metabolism, with potential minor contribution from CYP2C19. Padsevonil was shown to be a time-dependent CYP2C19-inhibitor, weak CYP3A4-inducer, weak inhibitor of P-gp/OCT1/MATE2-K, and potent OCT2-inhibitor. Initial clinical pharmacology studies in healthy participants showed that padsevonil had (i) good absorption, (ii) clearance mediated mainly by metabolism, and (iii) time-dependent kinetics. A study in genotyped participants confirmed the role of CYP2C19 in clearance and time-dependent kinetics; the major contribution of CYP3A4 was confirmed in DDI studies with CYP3A4-inducers (carbamazepine, oxcarbazepine) and -inhibitor (erythromycin). Padsevonil did not affect pharmacokinetics of valproate/lamotrigine/levetiracetam/oxcarbazepine or oral contraceptives. In a cocktail clinical study, padsevonil showed moderate CYP2C19 inhibition (omeprazole) and weak CYP3A4 induction (oral midazolam). No specific effects on CYP1A2 (caffeine), CYP2C9 (S-warfarin), and CYP2D6 (dextromethorphan) were observed. CONCLUSIONS The studies presented helped in understanding padsevonil disposition and risks of DDIs, which would inform dosing and prescribing. CLINICAL TRIAL REGISTRATION https://www.clinicaltrials.gov identifiers are NCT04131517, NCT03480243, NCT03695094, NCT04075409.
Collapse
|
7
|
Rollison HE, Mitra P, Chanteux H, Fang Z, Liang X, Park SH, Costales C, Hanna I, Thakkar N, Vergis JM, Bow DAJ, Hillgren KM, Brumm J, Chu X, Hop CECA, Lai Y, Li CY, Mahar KM, Salphati L, Sane R, Shen H, Taskar K, Taub M, Tohyama K, Xu C, Fenner KS. Survey of Pharmaceutical Industry's Best Practices around In Vitro Transporter Assessment and Implications for Drug Development: Considerations from the International Consortium for Innovation and Quality for Pharmaceutical Development Transporter Working Group. Drug Metab Dispos 2024; 52:582-596. [PMID: 38697852 DOI: 10.1124/dmd.123.001587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024] Open
Abstract
The International Consortium for Innovation and Quality in Pharmaceutical Development Transporter Working Group had a rare opportunity to analyze a crosspharma collation of in vitro data and assay methods for the evaluation of drug transporter substrate and inhibitor potential. Experiments were generally performed in accordance with regulatory guidelines. Discrepancies, such as not considering the impact of preincubation for inhibition and free or measured in vitro drug concentrations, may be due to the retrospective nature of the dataset and analysis. Lipophilicity was a frequent indicator of crosstransport inhibition (P-gp, BCRP, OATP1B, and OCT1), with high molecular weight (MW ≥500 Da) also common for OATP1B and BCRP inhibitors. A high level of overlap in in vitro inhibition across transporters was identified for BCRP, OATP1B1, and MATE1, suggesting that prediction of DDIs for these transporters will be common. In contrast, inhibition of OAT1 did not coincide with inhibition of any other transporter. Neutrals, bases, and compounds with intermediate-high lipophilicity tended to be P-gp and/or BCRP substrates, whereas compounds with MW <500 Da tended to be OAT3 substrates. Interestingly, the majority of in vitro inhibitors were not reported to be followed up with a clinical study by the submitting company, whereas those compounds identified as substrates generally were. Approaches to metabolite testing were generally found to be similar to parent testing, with metabolites generally being equally or less potent than parent compounds. However, examples where metabolites inhibited transporters in vitro were identified, supporting the regulatory requirement for in vitro testing of metabolites to enable integrated clinical DDI risk assessment. SIGNIFICANCE STATEMENT: A diverse dataset showed that transporter inhibition often correlated with lipophilicity and molecular weight (>500 Da). Overlapping transporter inhibition was identified, particularly that inhibition of BCRP, OATP1B1, and MATE1 was frequent if the compound inhibited other transporters. In contrast, inhibition of OAT1 did not correlate with the other drug transporters tested.
Collapse
Affiliation(s)
- Helen E Rollison
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Pallabi Mitra
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Hugues Chanteux
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Zhizhou Fang
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Xiaomin Liang
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Seong Hee Park
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Chester Costales
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Imad Hanna
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Nilay Thakkar
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - James M Vergis
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Daniel A J Bow
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Kathleen M Hillgren
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Jochen Brumm
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Xiaoyan Chu
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Cornelis E C A Hop
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Yurong Lai
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Cindy Yanfei Li
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Kelly M Mahar
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Laurent Salphati
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Rucha Sane
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Hong Shen
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Kunal Taskar
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Mitchell Taub
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Kimio Tohyama
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Christine Xu
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| | - Katherine S Fenner
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (H.E.R., K.S.F.); Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (P.M., M.T.); Quantitative Clinical Pharmacology, Development Sciences, UCB Biopharma SRL, Braine-L'Alleud, Belgium (H.C.); NCE Drug Metabolism and Pharmacokinetics, the healthcare business of Merck KGaA, Darmstadt, Germany (Z.F.); Drug Metabolism, Gilead Sciences, Inc. Foster City, California (X.L., Y.L.); Preclinical Sciences and Translational Safety, Janssen R&D LLC, Spring House, Pennsylvania (S.H.P.); Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut (C.C.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, East Hanover, New Jersey (I.H.); Clinical Pharmacology Modelling and Simulations, GlaxoSmithKline Research and Development, Collegeville, Pennsylvania (N.T., K.M.M.); IQ Secretariat, Faegre Drinker Biddle & Reath, LLP., Washington DC (J.M.V.); Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois (D.A.J.B.); Investigative Drug Disposition, Lilly Research Laboratories, Eli Lilly Inc, Indianapolis, Indiana (K.M.H.); Nonclinical Biostatistics, Genentech, Inc., South San Francisco, California (J.B.); ADME and Discovery Toxicity, Merck & Co., Inc., Rahway, New Jersey (X.C.); Departments of Drug Metabolism and Pharmacokinetics (C.E.C.A.H., L.S.) and Clinical Pharmacology (R.S.), Genentech, Inc., South San Francisco, California; Department of Pharmacokinetics and Drug Metabolism, Amgen Inc. South San Francisco, California (C.Y.L.); Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey (H.S.); DMPK Modeling, IVIVT, Research, GSK, Stevenage, United Kingdom (Ku.T.); Takeda Pharmaceutical Company Limited, Fujisawa, Japan (Ki.T.); and Pharmacokinetics, Dynamics, and Metabolism, Translational Medicine and Early Development, Sanofi US, Bridgewater, NJ (C.X.)
| |
Collapse
|
8
|
Xu Q, Sang Y, Gao A, Li L. The effects of drug-drug interaction on linezolid pharmacokinetics: A systematic review. Eur J Clin Pharmacol 2024; 80:785-795. [PMID: 38421436 DOI: 10.1007/s00228-024-03652-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/10/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVES Linezolid is a commonly used antibiotic in the clinical treatment of gram-positive bacterial infections. The impacts of drug interactions on the pharmacokinetics of linezolid are often overlooked. This manuscript aims to review the medications that affect the pharmacokinetics of linezolid. METHODS In accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we queried the PubMed, Embase, and Cochrane Library for publications from database establishment to November 3, 2023, using the search terms: "Linezolid" and "interaction," or "interact," or "drug-drug interaction," or "co-treatment," or "cotreatment," or "combined," or "combination." RESULTS A total of 24 articles were included. Among the reported medication interactions, rifampicin, levothyroxine, venlafaxine, and phenobarbital could reduce the concentration of linezolid; clarithromycin, digoxin, cyclosporine, proton pump inhibitors, and amiodarone could increase the concentration of linezolid, while aztreonam, phenylpropanolamine, dextromethorphan, antioxidant vitamins, and magnesium-containing antacids had no significant effects on linezolid pharmacokinetics. The ratio of mean (ROM) of linezolid AUC in co-treatment with rifampicin to monotherapy was 0.67 (95%CI 0.58-0.77) and 0.63 (95%CI 0.43-0.91), respectively, in 2 studies, and co-treatment with 500 mg clarithromycin to monotherapy was 1.81 (95%CI 1.49-2.13). CONCLUSIONS This systematic review found that numerous drugs have an impact on the pharmacokinetics of linezolid, and the purported main mechanism may be that linezolid is the substrate of P-glycoprotein. In clinical practice, it is prudent to pay attention to the changes in linezolid pharmacokinetics caused by interactions. Conducting therapeutic drug monitoring (TDM) is beneficial to improve efficacy and reduce adverse reactions of linezolid.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine for Clinical Evaluation and Translational Research, Hangzhou, China
| | - Yanlei Sang
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anna Gao
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lu Li
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine for Clinical Evaluation and Translational Research, Hangzhou, China.
| |
Collapse
|
9
|
Choi H, Huang F, Flack M. The Effect of BI 730357 (Retinoic Acid-Related Orphan Receptor Gamma t Antagonist, Bevurogant) on the Pharmacokinetics of a Transporter Probe Cocktail, Including Digoxin, Furosemide, Metformin, and Rosuvastatin: An Open-Label, Non-randomized, 2-Period Fixed-Sequence Trial in Healthy Subjects. Clin Pharmacol Drug Dev 2024; 13:197-207. [PMID: 37960990 DOI: 10.1002/cpdd.1344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023]
Abstract
Evaluating Drug-Drug Interactions (DDIs) for new investigational compounds requires several trials evaluating different drugs with different transporter specificities. By using a cocktail of drugs with different transporter specificities, a single trial could evaluate the pharmacokinetics (PKs) of each cocktail drug simultaneously, reducing the number of clinical DDI trials required for clinical development. We aimed to investigate the effect of steady-state Boehringer Ingelheim (BI) 730357 (bevurogant) on the PKs of a validated and optimized 4-component transporter cocktail. This open-label, non-randomized, 2-period fixed-sequence phase I trial compared transporter cocktail (0.25 mg digoxin/1 mg furosemide/10 mg metformin hydrochloride/10 mg rosuvastatin) with and without BI 730357 in healthy subjects aged 18-55 years with body mass index 18.5-29.9 kg/m2 . During reference treatment/period 1, transporter cocktail was administered 90 minutes after breakfast. After a washout period, during test treatment/period 2, BI 730357 was dosed twice daily for 13 days, with transporter cocktail administered on day 1. The primary endpoints were the area under the concentration-time curve of the analyte in plasma over the time interval from 0 extrapolated to infinity (AUC0-∞ ) and the maximum measured concentration of the analyte in plasma (Cmax ), and the secondary endpoint was the area under the concentration-time curve of the analyte in plasma over the time interval from 0 to the last quantifiable data point (AUC0-tz ). Steady-state BI 730357 increased digoxin (+48% to +94%), minimally affected metformin (-2% to -9%), furosemide (+12% to +18%), and rosuvastatin (+19% to +39%) exposure. Therefore, no clinically relevant inhibition of transporters OCT2/MATE-1/MATE-2K, OAT1/OAT3, OATP1B1/OATP1B3 was observed. Potential inhibition of breast cancer resistance protein noted as PK parameters of coproporphyrin I/III (OATP1B1/OATP1B3 biomarkers) remained within bioequivalence boundaries while rosuvastatin PK parameters (AUC0-∞ /Cmax /AUC0-tz ) exceeded the bioequivalence boundary. BI 730357 was safe and well tolerated. This trial confirms the usefulness and tolerability of the transporter cocktail consisting of digoxin, furosemide, metformin, and rosuvastatin in assessing drug-transporter interactions in vivo.
Collapse
Affiliation(s)
- HeeJae Choi
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Fenglei Huang
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Mary Flack
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| |
Collapse
|
10
|
Maeda K. Quantitative Prediction of Intestinal Absorption of Drugs from In Vitro Study: Utilization of Differentiated Intestinal Epithelial Cells Derived from Intestinal Stem Cells at Crypts. Drug Metab Dispos 2023; 51:1136-1144. [PMID: 37142427 DOI: 10.1124/dmd.122.000966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 03/21/2023] [Accepted: 04/28/2023] [Indexed: 05/06/2023] Open
Abstract
Prediction of intestinal absorption of drugs in humans is one of the critical elements in the development process for oral drugs. However, it remains challenging, because intestinal absorption of drugs is influenced by multiple factors, including the function of various metabolic enzymes and transporters, and large species differences in drug bioavailability hinder the prediction of human bioavailability directly from in vivo animal experiments. For the screening of intestinal absorption properties of drugs, a transcellular transport assay with Caco-2 cells is still routinely used by pharmaceutical companies because of its convenience, but the predictability of the fraction of the oral dose that goes to the portal vein of metabolic enzyme/transporter substrate drugs was not always good because the cellular expression of metabolic enzymes and transporters is different from that in the human intestine. Recently, various novel in vitro experimental systems have been proposed such as the use of human-derived intestinal samples, transcellular transport assay with induced pluripotent stem-derived enterocyte-like cells, or differentiated intestinal epithelial cells derived from intestinal stem cells at crypts. Crypt-derived differentiated epithelial cells have an excellent potential to characterize species differences and regional differences in intestinal absorption of drugs because a unified protocol can be used for the proliferation of intestinal stem cells and their differentiation into intestinal absorptive epithelial cells regardless of the animal species and the gene expression pattern of differentiated cells is maintained at the site of original crypts. The advantages and disadvantages of novel in vitro experimental systems for characterizing intestinal absorption of drugs are also discussed. SIGNIFICANCE STATEMENT: Among novel in vitro tools for the prediction of human intestinal absorption of drugs, crypt-derived differentiated epithelial cells have many advantages. Cultured intestinal stem cells are rapidly proliferated and easily differentiated into intestinal absorptive epithelial cells simply by changing the culture media. A unified protocol can be used for the establishment of intestinal stem cell culture from preclinical species and humans. Region-specific gene expression at the collection site of crypts can be reproduced in differentiated cells.
Collapse
Affiliation(s)
- Kazuya Maeda
- Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan
| |
Collapse
|
11
|
Gagliardi A, Bajraktari-Sylejmani G, Barocelli E, Weiss J, Rigalli JP. Extracellular Vesicles as Surrogates for Drug Metabolism and Clearance: Promise vs. Reality. Life (Basel) 2023; 13:1745. [PMID: 37629602 PMCID: PMC10455864 DOI: 10.3390/life13081745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Drug-metabolizing enzymes (DMEs) and transporters play a major role in drug efficacy and safety. They are regulated at multiple levels and by multiple factors. Estimating their expression and activity could contribute to predicting drug pharmacokinetics and their regulation by drugs or pathophysiological situations. Determining the expression of these proteins in the liver, intestine, and kidney requires the collection of biopsy specimens. Instead, the isolation of extracellular vesicles (EVs), which are nanovesicles released by most cells and present in biological fluids, could deliver this information in a less invasive way. In this article, we review the use of EVs as surrogates for the expression and activity of DMEs, uptake, and efflux transporters. Preliminary evidence has been provided for a correlation between the expression of some enzymes and transporters in EVs and the tissue of origin. In some cases, data obtained in EVs reflect the induction of phase I-DMEs in the tissues. Further studies are required to elucidate to what extent the regulation of other DMEs and transporters in the tissues reflects in the EV cargo. If an association between tissues and their EVs is firmly established, EVs may represent a significant advancement toward precision therapy based on the biotransformation and excretion capacity of each individual.
Collapse
Affiliation(s)
- Anna Gagliardi
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Gzona Bajraktari-Sylejmani
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Elisabetta Barocelli
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Juan Pablo Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
12
|
Translatability of in vitro Inhibition Potency to in vivo P-Glycoprotein Mediated Drug Interaction Risk. J Pharm Sci 2023; 112:1715-1723. [PMID: 36682487 DOI: 10.1016/j.xphs.2023.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023]
Abstract
P-glycoprotein (P-gp) may limit oral drug absorption of substrate drugs due to intestinal efflux. Therefore, regulatory agencies require investigation of new chemical entities as possible inhibitors of P-gp in vitro. Unfortunately, inter-laboratory and inter-assay variability have hindered the translatability of in vitro P-gp inhibition data to predict clinical drug interaction risk. The current study was designed to evaluate the impact of potential IC50 discrepancies between two commonly utilized assays, i.e., bi-directional Madin-Darby Canine Kidney-MDR1 cell-based and MDR1 membrane vesicle-based assays. When comparing vesicle- to cell-based IC50 values (n = 28 inhibitors), non-P-gp substrates presented good correlation between assay formats, whereas IC50s of P-gp substrates were similar or lower in the vesicle assays. The IC50s obtained with a cell line expressing relatively low P-gp aligned more closely to those obtained from the vesicle assay, but passive permeability of the inhibitors did not appear to influence the correlation of IC50s, suggesting that efflux activity reduces intracellular inhibitor concentrations. IC50s obtained between two independent laboratories using the same assay type showed good correlation. Using the G-value (i.e., ratio of estimated gut concentration-to-inhibition potency) >10 cutoff recommended by regulatory agencies resulted in minimal differences in predictive performance, suggesting this cutoff is appropriate for either assay format.
Collapse
|
13
|
Taskar KS, Yang X, Neuhoff S, Patel M, Yoshida K, Paine MF, Brouwer KL, Chu X, Sugiyama Y, Cook J, Polli JW, Hanna I, Lai Y, Zamek-Gliszczynski M. Clinical Relevance of Hepatic and Renal P-gp/BCRP Inhibition of Drugs: An International Transporter Consortium Perspective. Clin Pharmacol Ther 2022; 112:573-592. [PMID: 35612761 PMCID: PMC9436425 DOI: 10.1002/cpt.2670] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/16/2022] [Indexed: 12/11/2022]
Abstract
The role of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in drug-drug interactions (DDIs) and limiting drug absorption as well as restricting the brain penetration of drugs with certain physicochemical properties is well known. P-gp/BCRP inhibition by drugs in the gut has been reported to increase the systemic exposure to substrate drugs. A previous International Transporter Consortium (ITC) perspective discussed the feasibility of P-gp/BCRP inhibition at the blood-brain barrier and its implications. This ITC perspective elaborates and discusses specifically the hepatic and renal P-gp/BCRP (referred as systemic) inhibition of drugs and whether there is any consequence for substrate drug disposition. This perspective summarizes the clinical evidence-based recommendations regarding systemic P-gp and BCRP inhibition of drugs with a focus on biliary and active renal excretion pathways. Approaches to assess the clinical relevance of systemic P-gp and BCRP inhibition in the liver and kidneys included (i) curation of DDIs involving intravenously administered substrates or inhibitors; (ii) in vitro-to-in vivo extrapolation of P-gp-mediated DDIs at the systemic level; and (iii) curation of drugs with information available about the contribution of biliary excretion and related DDIs. Based on the totality of evidence reported to date, this perspective supports limited clinical DDI risk upon P-gp or BCRP inhibition in the liver or kidneys.
Collapse
Affiliation(s)
- Kunal S. Taskar
- Drug Metabolism and Pharmacokinetics, IVIVT, GlaxoSmithKline, Stevenage, UK
| | - Xinning Yang
- Office of Clinical Pharmacology, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Sibylle Neuhoff
- Certara UK Ltd, Simcyp Division, 1 Concourse Way, Level 2-Acero, Sheffield, S1 2BJ, UK
| | - Mitesh Patel
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kenta Yoshida
- Clinical Pharmacology, Genentech Early Research and Development, South San Francisco, CA 94080, USA
| | - Mary F. Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA
| | - Kim L.R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Xiaoyan Chu
- Department of ADME and Discovery Toxicology, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033 USA
| | - Yuichi Sugiyama
- Laboratory of Quantitative System PK/Pharmacodynamics, School of Pharmacy, Kioicho campus, Josai International University, Tokyo 102-0093, Japan
| | - Jack Cook
- Clinical Pharmacology, Global Product Development, Pfizer Inc., Groton, Connecticut, USA
| | - Joseph W. Polli
- Global Medical Sciences, ViiV Healthcare, Research Triangle Park NC USA
| | - Imad Hanna
- Pharmacokinetic Sciences-Oncology, Novartis Institute for Biomedical Research, East Hanover, NJ
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc. Foster City, CA USA
| | | |
Collapse
|
14
|
Yabut J, Houle R, Wang S, Liaw A, Katwaru R, Collier H, Hittle L, Chu X. Selection of an Optimal In Vitro Model to Assess P-gp Inhibition: Comparison of Vesicular and Bidirectional Transcellular Transport Inhibition Assays. Drug Metab Dispos 2022; 50:909-922. [PMID: 35489778 DOI: 10.1124/dmd.121.000807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/04/2022] [Indexed: 11/22/2022] Open
Abstract
The multidrug resistance protein 1 (MDR1) P-glycoprotein (P-gp) is a clinically important transporter. In vitro P-gp inhibition assays have been routinely conducted to predict the potential for clinical drug-drug interactions (DDIs) mediated by P-gp. However, high interlaboratory and intersystem variability of P-gp IC50 data limits accurate prediction of DDIs using static models and decision criteria recommended by regulatory agencies. In this study, we calibrated two in vitro P-gp inhibition models: vesicular uptake of N-methyl-quinidine (NMQ) in MDR1 vesicles and bidirectional transport (BDT) of digoxin in Lilly Laboratories Cell Porcine Kidney 1 cells overexpressing MDR1 (LLC-MDR1) using a total of 48 P-gp inhibitor and noninhibitor drugs and digoxin DDI data from 70 clinical studies. Refined thresholds were derived using receiver operating characteristic analysis, and their predictive performance was compared with the decision frameworks proposed by regulatory agencies and selected reference. Furthermore, the impact of various IC50 calculation methods and nonspecific binding of drugs on DDI prediction was evaluated. Our studies suggest that the concentration of inhibitor based on highest approved dose dissolved in 250 ml divided by IC50(I2/IC50) is sufficient to predict P-gp related intestinal DDIs. IC50 obtained from vesicular inhibition assay with a refined threshold of I2/IC50 ≥ 25.9 provides comparable predictive power over those measured by net secretory flux and efflux ratio in LLC-MDR1 cells. We therefore recommend vesicular P-gp inhibition as our preferred method given its simplicity, lower variability, higher assay throughput, and more direct estimation of in vitro kinetic parameters, rather than BDT assay. SIGNIFICANCE STATEMENT: This study has conducted comprehensive calibration of two in vitro P-gp inhibition models: uptake in MDR1 vesicles and bidirectional transport in LLC-MDR1 cell monolayers to predict DDIs. This study suggests that IC50s obtained from vesicular inhibition with a refined threshold of I2/IC50 ≥ 25.9 provide comparable predictive power over those in LLC-MDR1 cells. Therefore, vesicular P-gp inhibition is recommended as the preferred method given its simplicity, lower variability, higher assay throughput, and more direct estimation of in vitro kinetic parameters.
Collapse
Affiliation(s)
- Jocelyn Yabut
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Robert Houle
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Shubing Wang
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Andy Liaw
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Ravi Katwaru
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Hannah Collier
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Lucinda Hittle
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (J.Y., R.H., R.K., H.C., L.H., X.C.), and Department of Biometrics Research (S.W., A.L.), Merck & Co., Inc., Kenilworth, New Jersey
| |
Collapse
|
15
|
Mar PL, Horbal P, Chung MK, Dukes JW, Ezekowitz M, Lakkireddy D, Lip GYH, Miletello M, Noseworthy PA, Reiffel JA, Tisdale JE, Olshansky B, Gopinathannair R. Drug Interactions Affecting Antiarrhythmic Drug Use. Circ Arrhythm Electrophysiol 2022; 15:e007955. [PMID: 35491871 DOI: 10.1161/circep.121.007955] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antiarrhythmic drugs (AAD) play an important role in the management of arrhythmias. Drug interactions involving AAD are common in clinical practice. As AADs have a narrow therapeutic window, both pharmacokinetic as well as pharmacodynamic interactions involving AAD can result in serious adverse drug reactions ranging from arrhythmia recurrence, failure of device-based therapy, and heart failure, to death. Pharmacokinetic drug interactions frequently involve the inhibition of key metabolic pathways, resulting in accumulation of a substrate drug. Additionally, over the past 2 decades, the P-gp (permeability glycoprotein) has been increasingly cited as a significant source of drug interactions. Pharmacodynamic drug interactions involving AADs commonly involve additive QT prolongation. Amiodarone, quinidine, and dofetilide are AADs with numerous and clinically significant drug interactions. Recent studies have also demonstrated increased morbidity and mortality with the use of digoxin and other AAD which interact with P-gp. QT prolongation is an important pharmacodynamic interaction involving mainly Vaughan-Williams class III AAD as many commonly used drug classes, such as macrolide antibiotics, fluoroquinolone antibiotics, antipsychotics, and antiemetics prolong the QT interval. Whenever possible, serious drug-drug interactions involving AAD should be avoided. If unavoidable, patients will require closer monitoring and the concomitant use of interacting agents should be minimized. Increasing awareness of drug interactions among clinicians will significantly improve patient safety for patients with arrhythmias.
Collapse
Affiliation(s)
- Philip L Mar
- Department of Medicine, Division of Cardiology, St. Louis University, St. Louis, MO (P.L.M., P.H.)
| | - Piotr Horbal
- Department of Medicine, Division of Cardiology, St. Louis University, St. Louis, MO (P.L.M., P.H.)
| | - Mina K Chung
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute (M.K.C.), Cleveland Clinic, OH
| | | | - Michael Ezekowitz
- Lankenau Heart Institute, Bryn Mawr Hospital & Sidney Kimmel Medical College (M.E.)
| | | | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool & Liverpool Heart & Chest Hospital, Liverpool, United Kingdom (G.Y.H.L.).,Department of Clinical Medicine, Aalborg, Denmark (G.Y.H.L.)
| | | | - Peter A Noseworthy
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (P.A.N.)
| | - James A Reiffel
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.)
| | - James E Tisdale
- College of Pharmacy, Purdue University (J.E.T.).,School of Medicine, Indiana University, Indianapolis (J.E.T.)
| | - Brian Olshansky
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City (B.O.)
| | | | | |
Collapse
|
16
|
Michiba K, Maeda K, Shimomura O, Miyazaki Y, Hashimoto S, Oda T, Kusuhara H. Usefulness of Human Jejunal Spheroid-Derived Differentiated Intestinal Epithelial Cells for the Prediction of Intestinal Drug Absorption in Humans. Drug Metab Dispos 2022; 50:204-213. [PMID: 34992074 DOI: 10.1124/dmd.121.000796] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/02/2022] [Indexed: 11/22/2022] Open
Abstract
This study aimed to demonstrate the usefulness of human jejunal spheroid-derived differentiated intestinal epithelial cells as a novel in vitro model for clarifying the impact of intestinal drug-metabolizing enzymes and transporters on the intestinal absorption of substrate drugs in humans. Three-dimensional human intestinal spheroids were successfully established from surgical human jejunal specimens and expanded for a long period using L-WRN-conditioned medium, which contains Wnt3a, R-spondin 3, and noggin. The mRNA expression levels of intestinal pharmacokinetics-related genes in the human jejunal spheroid-derived differentiated intestinal epithelial cells were drastically increased over a 5-day period after seeding compared with those in human jejunal spheroids and were approximately the same as those in human jejunal tissue over a culture period of at least 13 days. Activities of typical drug-metabolizing enzymes [cytochrome P450 (CYP) 3A, CYP2C9, uridine 5'-diphospho-glucuronosyltransferase 1A, and carboxylesterase 2] and uptake/efflux transporters [peptide transporter 1/solute carrier 15A1], P-glycoprotein, and breast cancer resistance protein) in the differentiated cells were confirmed. Furthermore, intestinal availability (Fg) values estimated from the apical-to-basolateral permeation clearance across cell monolayer showed a good correlation with the in vivo Fg values in humans for five CYP3A substrate drugs (Fg range, 0.35-0.98). In conclusion, the functions of major intestinal drug-metabolizing enzymes and transporters could be maintained in human jejunal spheroid-derived differentiated intestinal epithelial cells. This model would be useful for the quantitative evaluation of the impact of intestinal drug-metabolizing enzymes and transporters on the intestinal absorption of substrate drugs in humans. SIGNIFICANCE STATEMENT: Limited information is available regarding the quantitative prediction of the impact of drug-metabolizing enzymes and transporters on the human intestinal absorption of substrates using in vitro assays with differentiated cells derived from human intestinal spheroids/organoids. This study confirmed the functions of typical drug-metabolizing enzymes and transporters in human jejunal spheroid-derived differentiated intestinal epithelial cells and demonstrated that intestinal availability (Fg) estimated from apical-to-basolateral permeation clearance across cell monolayers showed a good correlation with in vivo human Fg for CYP3A substrates.
Collapse
Affiliation(s)
- Kazuyoshi Michiba
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan (K.Ma.); and Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (O.S., Y.M., S.H., T.O.)
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan (K.Ma.); and Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (O.S., Y.M., S.H., T.O.)
| | - Osamu Shimomura
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan (K.Ma.); and Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (O.S., Y.M., S.H., T.O.)
| | - Yoshihiro Miyazaki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan (K.Ma.); and Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (O.S., Y.M., S.H., T.O.)
| | - Shinji Hashimoto
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan (K.Ma.); and Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (O.S., Y.M., S.H., T.O.)
| | - Tatsuya Oda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan (K.Ma.); and Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (O.S., Y.M., S.H., T.O.)
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan (K.Ma.); and Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (O.S., Y.M., S.H., T.O.)
| |
Collapse
|
17
|
Kurosawa K, Noguchi S, Nishimura T, Tomi M, Chiba K. Transplacental Pharmacokinetic Model of Digoxin Based on Ex Vivo Human Placental Perfusion Study. Drug Metab Dispos 2022; 50:287-298. [PMID: 34903589 DOI: 10.1124/dmd.121.000648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/10/2021] [Indexed: 11/22/2022] Open
Abstract
Digoxin is used as first-line therapy to treat fetal supraventricular tachycardia; however, because of the narrow therapeutic window, it is essential to estimate digoxin exposure in the fetus. The data from ex vivo human placental perfusion study are used to predict in vivo fetal exposure noninvasively, but the ex vivo fetal-to-maternal concentration (F:M) ratios observed in digoxin perfusion studies were much lower than those in vivo. In the present study, we developed a human transplacental pharmacokinetic model of digoxin using previously reported ex vivo human placental perfusion data. The model consists of maternal intervillous, fetal capillary, non-perfused tissue, and syncytiotrophoblast compartments, with multidrug resistance protein (MDR) 1 and influx transporter at the microvillous membrane (MVM) and influx and efflux transporters at the basal plasma membrane (BM). The model-predicted F:M ratio was 0.66, which is consistent with the mean in vivo value of 0.77 (95% confidence interval: 0.64-0.91). The time to achieve the steady state from the ex vivo perfusion study was estimated as 1,500 minutes, which is considerably longer than the reported ex vivo experimental durations, and this difference is considered to account for the inconsistency between ex vivo and in vivo F:M ratios. Reported digoxin concentrations in a drug-drug interaction study with MDR1 inhibitors quinidine and verapamil were consistent with the profiles simulated by our model incorporating inhibition of efflux transporter at the BM in addition to MVM. Our modeling and simulation approach should be a powerful tool to predict fetal exposure and DDIs in human placenta. SIGNIFICANCE STATEMENT: We developed a human transplacental pharmacokinetic model of digoxin based on ex vivo human placental perfusion studies in order to resolve inconsistencies between reported ex vivo and in vivo fetal-to-maternal concentration ratios. The model successfully predicted the in vivo fetal exposure to digoxin and the drug-drug interactions of digoxin and P-glycoprotein/multidrug resistance protein 1 inhibitors in human placenta.
Collapse
Affiliation(s)
- Ken Kurosawa
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Saki Noguchi
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Tomohiro Nishimura
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Masatoshi Tomi
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Koji Chiba
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| |
Collapse
|
18
|
Hirai T, Kasai H, Naganuma M, Hagiwara N, Shiga T. Population pharmacokinetic analysis and dosage recommendations for digoxin in Japanese patients with atrial fibrillation and heart failure using real-world data. BMC Pharmacol Toxicol 2022; 23:14. [PMID: 35144695 PMCID: PMC8830040 DOI: 10.1186/s40360-022-00552-y] [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: 09/07/2021] [Accepted: 01/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Digoxin is an important treatment option for reducing the ventricular rate in patients with atrial fibrillation (AF) and heart failure (HF). Digoxin has a narrow therapeutic window and large interindividual variability. A low target blood concentration, especially ≤0.9 ng/mL, is recommended for patients with HF who are taking digoxin. This study aimed to develop a population pharmacokinetic model and to identify clinical factors that affect digoxin exposure and an optimal digoxin dosing regimen in Japanese patients with AF and HF. METHODS A population pharmacokinetic analysis was performed by using a nonlinear mixed effects model based on 3465 concentration points from 391 patients (>18 years) who were receiving oral digoxin. Using trough serum digoxin concentrations and clinical data, a population pharmacokinetic model was developed for determining covariates of clearance. A 1-compartment model was used to examine the interindividual variability of the oral clearance (CL/F) of digoxin. An appropriate dosage of digoxin was identified using Monte Carlo simulation. RESULTS The final model demonstrated that creatinine clearance (CLCR) and the use of amiodarone were factors that contributed to the CL/F of digoxin. Monte Carlo simulation results showed that with a daily maintenance dose of 0.25 mg, the intoxication risk window of a trough serum concentration of ≥0.9 ng/mL could be reached in more than half of patients regardless of renal function category or concurrent use of amiodarone. The appropriate maintenance dosage was 0.125 mg daily for most Japanese patients with AF and HF. However, with a daily dose of 0.125 mg, a trough serum concentration of ≥0.9 ng/mL could be reached in more than half of patients with renal impairments (CLCR 30 mL/min) or concurrent use of amiodarone. A daily maintenance dose of 0.0625 mg was acceptable for these patients. CONCLUSIONS CLCR and the use of amiodaron were found to contribute to digoxin clearance using a population pharmacokinetic methodology. For Japanese patients with AF and HF, 0.125 mg is an appropriate daily digoxin maintenance dose, but a dose reduction is required for patients with CLCR <30 mL/min or concurrent amiodarone use.
Collapse
Affiliation(s)
- Toshinori Hirai
- Department of Clinical Pharmacology and Therapeutics, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-8461, Japan.,Department of Pharmacy, Faculty of Medicine, Mie University Hospital, Mie University, Tsu, Japan
| | | | - Miyoko Naganuma
- Department of Pharmacy, International University of Health and Welfare Atami Hospital, Atami, Japan
| | - Nobuhisa Hagiwara
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Tsuyoshi Shiga
- Department of Clinical Pharmacology and Therapeutics, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-8461, Japan. .,Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
19
|
Saib S, Hodin S, Bin V, Ollier E, Delavenne X. In Vitro Evaluation of P-gp-Mediated Drug-Drug Interactions Using the RPTEC/TERT1 Human Renal Cell Model. Eur J Drug Metab Pharmacokinet 2021; 47:223-233. [PMID: 34935100 DOI: 10.1007/s13318-021-00744-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND OBJECTIVES In vitro evaluation of the P-glycoprotein (P-gp) inhibitory potential is an important issue when predicting clinically relevant drug-drug interactions (DDIs). Located within all physiological barriers, including intestine, liver, and kidneys, P-gp plays a major role in the pharmacokinetics of various therapeutic classes. However, few data are available about DDIs involving renal transporters during the active tubular secretion of drugs. In this context, the present study was designed to investigate the application of the human renal cell line RPTEC/TERT1 to study drug interactions mediated by P-gp. METHODS The P-gp inhibitory potentials of a panel of drugs were first determined by measuring the intracellular accumulation of rhodamine 123 in RPTEC/TERT1 cells. Then four drugs were selected to assess the half-maximal inhibitor concentration (IC50) values by measuring the intracellular accumulation of two P-gp-substrate drugs, apixaban and rivaroxaban. Finally, according to the FDA guidelines, the [I1]/IC50 ratio was calculated for each combination of drugs to assess the clinical relevance of the DDIs. RESULTS The data showed that drugs which are known P-gp inhibitors, including cyclosporin A, ketoconazole, and verapamil, caused great increases in rhodamine 123 retention, whereas noninhibitors did not affect the intracellular accumulation of the P-gp substrate. The determined IC50 values were in accordance with the inhibition profiles observed in the rhodamine 123 accumulation assays, confirming the reliability of the RPTEC/TERT1 model. CONCLUSIONS Taken together, the data demonstrate the feasibility of the application of the RPTEC/TERT1 model for evaluating the P-gp inhibitory potentials of drugs and consequently predicting renal drug interactions.
Collapse
Affiliation(s)
- Sonia Saib
- INSERM U1059, Dysfonction Vasculaire et Hémostase, Université Jean Monnet, 10 rue de la Marandière, Campus Santé Innovations, Saint-Priest-en-Jarez, Saint-Etienne, France.
| | - Sophie Hodin
- INSERM U1059, Dysfonction Vasculaire et Hémostase, Université Jean Monnet, 10 rue de la Marandière, Campus Santé Innovations, Saint-Priest-en-Jarez, Saint-Etienne, France
| | - Valérie Bin
- INSERM U1059, Dysfonction Vasculaire et Hémostase, Université Jean Monnet, 10 rue de la Marandière, Campus Santé Innovations, Saint-Priest-en-Jarez, Saint-Etienne, France
| | - Edouard Ollier
- INSERM U1059, Dysfonction Vasculaire et Hémostase, Université Jean Monnet, 10 rue de la Marandière, Campus Santé Innovations, Saint-Priest-en-Jarez, Saint-Etienne, France
| | - Xavier Delavenne
- INSERM U1059, Dysfonction Vasculaire et Hémostase, Université Jean Monnet, 10 rue de la Marandière, Campus Santé Innovations, Saint-Priest-en-Jarez, Saint-Etienne, France.,Laboratoire de Pharmacologie Toxicologie Gaz du sang, CHU de Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
20
|
Hashimoto Y, Michiba K, Maeda K, Kusuhara H. Quantitative prediction of pharmacokinetic properties of drugs in humans: Recent advance in in vitro models to predict the impact of efflux transporters in the small intestine and blood-brain barrier. J Pharmacol Sci 2021; 148:142-151. [PMID: 34924119 DOI: 10.1016/j.jphs.2021.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Efflux transport systems are essential to suppress the absorption of xenobiotics from the intestinal lumen and protect the critical tissues at the blood-tissue barriers, such as the blood-brain barrier. The function of drug efflux transport is dominated by various transporters. Accumulated clinical evidences have revealed that genetic variations of the transporters, together with coadministered drugs, affect the expression and/or function of transporters and subsequently the pharmacokinetics of substrate drugs. Thus, in the preclinical stage of drug development, quantitative prediction of the impact of efflux transporters as well as that of uptake transporters and metabolic enzymes on the pharmacokinetics of drugs in humans has been performed using various in vitro experimental tools. Various kinds of human-derived cell systems can be applied to the precise prediction of drug transport in humans. Mathematical modeling consisting of each intrinsic metabolic or transport process enables us to understand the disposition of drugs both at the organ level and at the level of the whole body by integrating a variety of experimental results into model parameters. This review focuses on the role of efflux transporters in the intestinal absorption and brain distribution of drugs, in addition to recent advances in predictive tools and methodologies.
Collapse
Affiliation(s)
- Yoshiki Hashimoto
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuyoshi Michiba
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuya Maeda
- Laboratory of Pharmaceutics, Kitasato University School of Pharmacy, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
21
|
Scheuenpflug J, Kropeit D, Erb-Zohar K, Theis JGW, Stobernack HP, McCormick D, Zimmermann H, Rübsamen-Schaeff H. The Effect of Oral Letermovir Administration on the Pharmacokinetics of a Single Oral Dose of P-Glycoprotein Substrate Digoxin in Healthy Volunteers. Clin Pharmacol Drug Dev 2021; 11:6-15. [PMID: 34812580 DOI: 10.1002/cpdd.1043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/27/2021] [Indexed: 11/12/2022]
Abstract
Letermovir is a human cytomegalovirus (CMV) terminase inhibitor approved in the United States, Canada, Japan, and the European Union for prophylaxis of CMV infection and disease in CMV-seropositive, allogeneic, hematopoietic stem-cell transplant recipients. In vitro, letermovir is a substrate and potential modulator of P-glycoprotein. The potential of letermovir to alter the pharmacokinetics of digoxin (a P-glycoprotein substrate) upon coadministration in healthy subjects was therefore investigated in a phase 1 trial (EudraCT: 2011-004516-39). Oral letermovir 240 mg was administered twice daily for 12 days with a single oral digoxin 0.5-mg dose on day 7; after a washout period, oral digoxin 0.5 mg was administered on day 35 (sequence 1). The period order was reversed after a 28-day washout for sequence 2. Pharmacokinetics and safety were evaluated. The presence of steady-state letermovir reduced digoxin area under the plasma concentration-time curve from administration until last quantifiable measurement by 12% and maximum plasma concentration by 22% compared with digoxin alone; digoxin half-life and elimination rate remained similar in both conditions. The between-subject variability of digoxin maximum plasma concentration was higher with letermovir than without (42% vs 31%) and similar for digoxin area under the plasma concentration-time curve in both periods. No specific safety or tolerability concerns were identified. Overall, letermovir had no clinically relevant effect on concomitant administration with digoxin.
Collapse
Affiliation(s)
- Jürgen Scheuenpflug
- AiCuris Anti-infective Cures AG, Wuppertal, Germany.,Merck KGaA, Darmstadt, Germany
| | - Dirk Kropeit
- AiCuris Anti-infective Cures AG, Wuppertal, Germany
| | | | | | | | - David McCormick
- AiCuris Anti-infective Cures AG, Wuppertal, Germany.,DMPK Solutions Ltd, Nottinghamshire, UK
| | | | | |
Collapse
|
22
|
Yamazaki S, Evers R, De Zwart L. Physiologically-based pharmacokinetic modeling to evaluate in vitro-to-in vivo extrapolation for intestinal P-glycoprotein inhibition. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 11:55-67. [PMID: 34668334 PMCID: PMC8752109 DOI: 10.1002/psp4.12733] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/12/2021] [Accepted: 10/12/2021] [Indexed: 11/08/2022]
Abstract
As one of the key components in model‐informed drug discovery and development, physiologically‐based pharmacokinetic (PBPK) modeling linked with in vitro‐to‐in vivo extrapolation (IVIVE) is widely applied to quantitatively predict drug–drug interactions (DDIs) on drug‐metabolizing enzymes and transporters. This study aimed to investigate an IVIVE for intestinal P‐glycoprotein (Pgp, ABCB1)‐mediated DDIs among three Pgp substrates, digoxin, dabigatran etexilate, and quinidine, and two Pgp inhibitors, itraconazole and verapamil, via PBPK modeling. For Pgp substrates, assuming unbound Michaelis‐Menten constant (Km) to be intrinsic, in vitro‐to‐in vivo scaling factors for maximal Pgp‐mediated efflux rate (Jmax) were optimized based on the clinically observed results without co‐administration of Pgp inhibitors. For Pgp inhibitors, PBPK models utilized the reported in vitro values of Pgp inhibition constants (Ki), 1.0 μM for itraconazole and 2.0 μM for verapamil. Overall, the PBPK modeling sufficiently described Pgp‐mediated DDIs between these substrates and inhibitors with the prediction errors of less than or equal to ±25% in most cases, suggesting a reasonable IVIVE for Pgp kinetics in the clinical DDI results. The modeling results also suggest that Pgp kinetic parameters of both the substrates (Km and Jmax) and the inhibitors (Ki) are sensitive to Pgp‐mediated DDIs, thus being key for successful DDI prediction. It would also be critical to incorporate appropriate unbound inhibitor concentrations at the site of action into PBPK models. The present results support a quantitative prediction of Pgp‐mediated DDIs using in vitro parameters, which will significantly increase the value of in vitro studies to design and run clinical DDI studies safely and effectively.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Drug Metabolism & Pharmacokinetics, Janssen Research & Development, LLC, San Diego, California, USA
| | - Raymond Evers
- Drug Metabolism & Pharmacokinetics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Loeckie De Zwart
- Drug Metabolism & Pharmacokinetics, Janssen Research & Development, Beerse, Belgium
| |
Collapse
|
23
|
Lang J, Vincent L, Chenel M, Ogungbenro K, Galetin A. Reduced physiologically-based pharmacokinetic model of dabigatran etexilate-dabigatran and its application for prediction of intestinal P-gp-mediated drug-drug interactions. Eur J Pharm Sci 2021; 165:105932. [PMID: 34260894 DOI: 10.1016/j.ejps.2021.105932] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/01/2021] [Accepted: 06/22/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Dabigatran etexilate (DABE) has been suggested as a clinical probe for intestinal P-glycoprotein (P-gp)-mediated drug-drug interaction (DDI) studies and, as an alternative to digoxin. Clinical DDI data with various P-gp inhibitors demonstrated a dose-dependent inhibition of P-gp with DABE. The aims of this study were to develop a joint DABE (prodrug)-dabigatran reduced physiologically-based-pharmacokinetic (PBPK) model and to evaluate its ability to predict differences in P-gp DDI magnitude between a microdose and a therapeutic dose of DABE. METHODS A joint DABE-dabigatran PBPK model was developed with a mechanistic intestinal model accounting for the regional P-gp distribution in the gastrointestinal tract. Model input parameters were estimated using DABE and dabigatran pharmacokinetic (PK) clinical data obtained after administration of DABE alone or with a strong P-gp inhibitor, itraconazole, and over a wide range of DABE doses (from 375 µg to 400 mg). Subsequently, the model was used to predict extent of DDI with additional P-gp inhibitors and with different DABE doses. RESULTS The reduced DABE-dabigatran PBPK model successfully described plasma concentrations of both prodrug and metabolite following administration of DABE at different dose levels and when co-administered with itraconazole. The model was able to capture the dose dependency in P-gp mediated DDI. Predicted magnitude of itraconazole P-gp DDI was higher at the microdose (predicted vs. observed median fold-increase in AUC+inh/AUCcontrol (min-max) = 5.88 (4.29-7.93) vs. 6.92 (4.96-9.66) ) compared to the therapeutic dose (predicted median fold-increase in AUC+inh/AUCcontrol = 3.48 (2.37-4.84) ). In addition, the reduced DABE-dabigatran PBPK model predicted successfully the extent of DDI with verapamil and clarithromycin as P-gp inhibitors. Model-based simulations of dose staggering predicted the maximum inhibition of P-gp when DABE microdose was concomitantly administered with itraconazole solution; simulations also highlighted dosing intervals required to minimise the DDI risk depending on the DABE dose administered (microdose vs. therapeutic). CONCLUSIONS This study provides a modelling framework for the evaluation of P-gp inhibitory potential of new molecular entities using DABE as a clinical probe. Simulations of dose staggering and regional differences in the extent of intestinal P-gp inhibition for DABE microdose and therapeutic dose provide model-based guidance for design of prospective clinical P-gp DDI studies.
Collapse
Affiliation(s)
- Jennifer Lang
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, United Kingdom
| | | | - Marylore Chenel
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, United Kingdom.
| |
Collapse
|
24
|
Classification of drugs for evaluating drug interaction in drug development and clinical management. Drug Metab Pharmacokinet 2021; 41:100414. [PMID: 34666290 DOI: 10.1016/j.dmpk.2021.100414] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 12/22/2022]
Abstract
During new drug development, clinical drug interaction studies are carried out in accordance with the mechanism of potential drug interactions evaluated by in vitro studies. The obtained information should be provided efficiently to medical experts through package inserts and various information materials after the drug's launch. A recently updated Japanese guideline presents general procedures that are considered scientifically valid at the present moment. In this review, we aim to highlight the viewpoints of the Japanese guideline and enumerate drugs that were involved or are anticipated to be involved in evident pharmacokinetic drug interactions and classify them by their clearance pathway and potential intensity based on systematic reviews of the literature. The classification would be informative for designing clinical studies during the development stage, and the appropriate management of drug interactions in clinical practice.
Collapse
|
25
|
Tsuchitani T, Akiyoshi T, Imaoka A, Ohtani H. Digoxin absorption decreased independently of P-gp activity in rats with irinotecan-induced gastrointestinal damage. J Pharm Health Care Sci 2021; 7:24. [PMID: 34193275 PMCID: PMC8247139 DOI: 10.1186/s40780-021-00207-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 04/26/2021] [Indexed: 11/15/2022] Open
Abstract
Background Irinotecan (CPT-11) is clinically known to cause severe diarrhea and gastrointestinal damage. Recently, we have reported that CPT-11-induced gastrointestinal damage is associated with the upregulation of intestinal P-glycoprotein (P-gp) expression and decreased absorption of its substrate, dabigatran etexilate (DABE), using a rat model. However, the P-gp activity or its contribution to the decreased absorption remains unclear. The aim of this study was to quantitatively evaluate how P-gp activity changes in rats with CPT-11-induced gastrointestinal damage, as assessed by the absorption of digoxin (DGX), a typical P-gp substrate. Methods Male Sprague-Dawley rats were intravenously administered CPT-11 at a dose of 60 mg/kg/day for 4 days to induce gastrointestinal damage. Then, the rats were administered DGX orally (40 μg/kg), after some of them were orally administered clarithromycin (CAM; 10 mg/kg), a P-gp inhibitor. DGX (30 μg/kg) was administered intravenously to determine the bioavailability (BA). The rats’ DGX plasma concentration profiles were determined using LC-MS/MS. Results CPT-11 treatment decreased the maximum concentration (Cmax) and area under the plasma concentration-time curve (AUCpo) of DGX, which does not contradict to the DABE study. Although in the CPT-11-treated group the BA of DGX was significantly decreased to 40% of the control value, CAM did not affect the BA of DGX in the CPT-11-treated group. Conclusions Increased P-gp expression in rats with CPT-11-induced gastrointestinal damage is not necessarily associated with increased P-gp activity or contribution to the drug absorption in vivo. The decreased DGX absorption observed in this study might be attributable to other factors, such as a reduction in the absorptive surface area of the gastrointestinal tract.
Collapse
Affiliation(s)
- Toshiaki Tsuchitani
- Division of Clinical Pharmacokinetics, Keio University Faculty of Pharmacy, 1-5-30, Shibakoen Minato-ku, Tokyo, 105-8512, Japan
| | - Takeshi Akiyoshi
- Division of Clinical Pharmacokinetics, Keio University Faculty of Pharmacy, 1-5-30, Shibakoen Minato-ku, Tokyo, 105-8512, Japan
| | - Ayuko Imaoka
- Division of Clinical Pharmacokinetics, Keio University Faculty of Pharmacy, 1-5-30, Shibakoen Minato-ku, Tokyo, 105-8512, Japan
| | - Hisakazu Ohtani
- Division of Clinical Pharmacokinetics, Keio University Faculty of Pharmacy, 1-5-30, Shibakoen Minato-ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
26
|
Martinec O, Biel C, de Graaf IAM, Huliciak M, de Jong KP, Staud F, Cecka F, Olinga P, Vokral I, Cerveny L. Rifampicin Induces Gene, Protein, and Activity of P-Glycoprotein (ABCB1) in Human Precision-Cut Intestinal Slices. Front Pharmacol 2021; 12:684156. [PMID: 34177592 PMCID: PMC8220149 DOI: 10.3389/fphar.2021.684156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/18/2021] [Indexed: 12/20/2022] Open
Abstract
P-glycoprotein (ABCB1), an ATP-binding cassette efflux transporter, limits intestinal absorption of its substrates and is a common site of drug–drug interactions. Drug-mediated induction of intestinal ABCB1 is a clinically relevant phenomenon associated with significantly decreased drug bioavailability. Currently, there are no well-established human models for evaluating its induction, so drug regulatory authorities provide no recommendations for in vitro/ex vivo testing drugs’ ABCB1-inducing activity. Human precision-cut intestinal slices (hPCISs) contain cells in their natural environment and express physiological levels of nuclear factors required for ABCB1 induction. We found that hPCISs incubated in William’s Medium E for 48 h maintained intact morphology, ATP content, and ABCB1 efflux activity. Here, we asked whether rifampicin (a model ligand of pregnane X receptor, PXR), at 30 μM, induces functional expression of ABCB1 in hPCISs over 24- and 48-h incubation (the time to allow complete induction to occur). Rifampicin significantly increased gene expression, protein levels, and efflux activity of ABCB1. Moreover, we described dynamic changes in ABCB1 transcript levels in hPCISs over 48 h incubation. We also observed that peaks of induction are achieved among donors at different times, and the extent of ABCB1 gene induction is proportional to PXR mRNA levels in the intestine. In conclusion, we showed that hPCISs incubated in conditions comparable to those used for inhibition studies can be used to evaluate drugs’ ABCB1-inducing potency in the human intestine. Thus, hPCISs may be valuable experimental tools that can be prospectively used in complex experimental evaluation of drug–drug interactions.
Collapse
Affiliation(s)
- Ondrej Martinec
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia.,Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Carin Biel
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Groningen, Netherlands
| | - Inge A M de Graaf
- Graduate School of Science, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Martin Huliciak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Koert P de Jong
- Department of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Filip Cecka
- Department of Surgery, University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Groningen, Netherlands
| | - Ivan Vokral
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Lukas Cerveny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| |
Collapse
|
27
|
Goda M, Kanda M, Yoshioka T, Yoshida A, Murai Y, Zamami Y, Aizawa F, Niimura T, Hamano H, Okada N, Yagi K, Chuma M, Izawa-Ishizawa Y, Ishizawa K. Effects of 5-HT ₃ receptor antagonists on cisplatin-induced kidney injury. Clin Transl Sci 2021; 14:1906-1916. [PMID: 33982438 PMCID: PMC8504842 DOI: 10.1111/cts.13045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/01/2022] Open
Abstract
Nausea, vomiting, and renal injury are the common adverse effects associated with cisplatin. Cisplatin is excreted via the multidrug and toxin release (MATE) transporter, and the involvement of the MATE transporter in cisplatin‐induced kidney injury has been reported. The MATE transporter is also involved in the excretion of ondansetron, but the effects of 5‐HT3 receptor antagonists used clinically for cisplatin‐induced renal injury have not been elucidated. Therefore, the aim of this study was to investigate the effects of 5‐HT3 receptor antagonists in a mouse model of cisplatin‐induced kidney injury and to validate the results using medical big data analysis of more than 1.4 million reports and a survey of 3000 hospital medical records. The concomitant use of a first‐generation 5‐HT3 receptor antagonist (ondansetron, granisetron, or ramosetron) significantly increased cisplatin accumulation in the kidneys and worsened renal damage. Conversely, the concomitant use of palonosetron had no effect on renal function compared with the use of cisplatin alone. Furthermore, an analysis of data from the US Food and Drug Administration Adverse Event Reporting System and retrospective medical records revealed that the combination treatment of cisplatin and a first‐generation 5‐HT3 receptor antagonist significantly increased the number of reported renal adverse events compared with the combination treatment of cisplatin and a second‐generation 5‐HT3 receptor antagonist. These results suggest that compared with the first‐generation antagonists, second‐generation 5‐HT3 receptor antagonists do not worsen cisplatin‐induced acute kidney injury. The findings should be validated in a prospective controlled trial before implementation in clinical practice.
Collapse
Affiliation(s)
- Mitsuhiro Goda
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan.,Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Masaya Kanda
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Toshihiko Yoshioka
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Ami Yoshida
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoichi Murai
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoshito Zamami
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Fuka Aizawa
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Takahiro Niimura
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hirofumi Hamano
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Naoto Okada
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Kenta Yagi
- Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Masayuki Chuma
- Department of Pharmacy, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| |
Collapse
|
28
|
Jeong HJ, Lee SH, Kang HE. Changes in digoxin pharmacokinetics associated with hepatic P-glycoprotein upregulation in rats with non-alcoholic fatty liver disease. Fundam Clin Pharmacol 2021; 35:1100-1108. [PMID: 33914974 DOI: 10.1111/fcp.12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND & OBJECTIVES Upregulation of hepatic P-glycoprotein (P-gp) expression has been reported in patients with non-alcoholic fatty liver disease (NAFLD) and rodent models thereof. Here, we explored the changes hepatic P-gp expression and activity in a NAFLD rat model and the effects thereof on the pharmacokinetics of digoxin (a probe substrate of P-gp). METHODS Rats were fed a 1% (w/w) orotic acid-containing diet for 20 days to induce NAFLD; control rats received a normal diet. P-gp expression and biliary digoxin excretion were examined. The pharmacokinetics of digoxin were evaluated after it had been administered intravenously (10 μg·kg-1 ) and orally (200 μg·kg-1 ) to control and NAFLD rats. RESULTS The total areas under the plasma concentration-time curves (AUCs) of digoxin after intravenous and oral administration were significantly smaller (by 39.1% and 73.0%, respectively) in NAFLD rats because of faster biliary digoxin excretion, reflecting elevations of hepatic P-gp expression and activity. Notably, the steady-state volume of distribution rose by 98.2%, while extent of oral bioavailability fell by 55.5% in NAFLD rats. CONCLUSION This is the first study to report digoxin pharmacokinetic changes caused by hepatic P-gp upregulation in NAFLD. Further studies are needed to explore the clinical impact of enhanced P-gp-mediated biliary excretion on pharmacotherapies using P-gp substrates in patients with NAFLD.
Collapse
Affiliation(s)
- Hee Jin Jeong
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, South Korea
| | - Song Hee Lee
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, South Korea
| | - Hee Eun Kang
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, South Korea
| |
Collapse
|
29
|
Tuey SM, Atilano-Roque A, Charkoftaki G, Thurman JM, Nolin TD, Joy MS. Influence of vitamin D treatment on functional expression of drug disposition pathways in human kidney proximal tubule cells during simulated uremia. Xenobiotica 2021; 51:657-667. [PMID: 33870862 DOI: 10.1080/00498254.2021.1909783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Effects of cholecalciferol (VitD3) and calcitriol (1,25-VitD3), on the expression and function of major vitamin D metabolizing enzymes (cytochrome P450 [CYP]2R1, CYP24A1) and select drug transport pathways (ABCB1/P-gp, SLCO4C1/OATP4C1) were evaluated in human kidney proximal tubule epithelial cells (hPTECs) under normal and uraemic serum conditions.hPTECs were incubated with 10% normal or uraemic serum for 24 h followed by treatment with 2% ethanol vehicle, or 100 and 240 nM doses of VitD3, or 1,25-VitD3 for 6 days. The effects of treatment on mRNA and protein expression and functional activity of select CYP enzymes and transporters were assessedUnder uraemic serum, treatment with 1,25-VitD3 resulted in increased mRNA but decreased protein expression of CYP2R1. Activity of CYP2R1 was not influenced by serum or VitD analogues. CYP24A1 expression was increased with 1,25-VitD3 under normal as well as uraemic serum, although to a lesser extent. ABCB1/P-gp mRNA expression increased under normal and uraemic serum, with exposure to 1,25-VitD3. SLCO4C1/OATP4C1 exhibited increased mRNA but decreased protein expression, under uraemic serum + 1,25-VitD3. Functional assessments of transport showed no changes regardless of exposure to serum or 1,25-VitD3.Key findings indicate that uraemic serum and VitD treatment led to differential effects on the functional expression of CYPs and transporters in hPTECs.
Collapse
Affiliation(s)
- Stacey M Tuey
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Amandla Atilano-Roque
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Georgia Charkoftaki
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA.,School of Public Health, Yale University, New Haven, CT, USA
| | - Joshua M Thurman
- Division of Nephrology and Hypertension, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Thomas D Nolin
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melanie S Joy
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA.,Division of Nephrology and Hypertension, School of Medicine, University of Colorado, Aurora, CO, USA
| |
Collapse
|
30
|
Kato T, Mikkaichi T, Yoshigae Y, Okudaira N, Shimizu T, Izumi T, Ando S, Matsumoto Y. Quantitative analysis of an impact of P-glycoprotein on edoxaban's disposition using a human physiologically based pharmacokinetic (PBPK) model. Int J Pharm 2021; 597:120349. [PMID: 33545293 DOI: 10.1016/j.ijpharm.2021.120349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/31/2020] [Accepted: 01/31/2021] [Indexed: 12/18/2022]
Abstract
The purpose of this study was to evaluate the impact of P-glycoprotein (P-gp) efflux on edoxaban absorption in gastrointestinal tracts quantitatively by a physiologically based pharmacokinetic (PBPK) model constructed with clinical and non-clinical observations (using GastroPlus™ software). An absorption process was described by the advanced compartmental absorption and transit model with the P-gp function. A human PBPK model was constructed by integrating the clinical and non-clinical observations. The constructed model was demonstrated to reproduce the data observed in the mass-balance study. Thus, elimination pathways can be quantitatively incorporated into the model. A constructed model successfully described the difference in slopes of plasma concentration (Cp)-time curve at around 8 - 24 hr post-dose between intravenous infusion and oral administration. Furthermore, the model without P-gp efflux activity can reproduce the Cp-time profile in the absence of P-gp activity observed from the clinical DDI study results. Since the difference of slopes between intravenous infusion and oral administration also disappeared by the absence of P-gp efflux activity, P-gp must be a key molecule to govern edoxaban's PK behavior. The constructed PBPK model will help us to understand the significant contribution of P-gp in edoxaban's disposition in gastrointestinal tracts quantitatively.
Collapse
Affiliation(s)
- Takafumi Kato
- Formulation Technology Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| | - Tsuyoshi Mikkaichi
- Drug Metabolism & Pharmacokinetics Research Laboratories,Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| | - Yasushi Yoshigae
- Drug Metabolism & Pharmacokinetics Research Laboratories,Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Noriko Okudaira
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. (Simcyp Division Certara, Inc., Tokyo, Japan), Tokyo, Japan
| | - Takako Shimizu
- Quantitative Clinical Pharmacology Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takashi Izumi
- Drug Metabolism & Pharmacokinetics Research Laboratories,Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Shuichi Ando
- Formulation Technology Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yoshiaki Matsumoto
- Laboratory of Clinical Pharmacokinetics, School of Pharmacy, Nihon University, Chiba, Japan
| |
Collapse
|
31
|
Ramzan M, Ahmed A, Usmani Y, Siddiqui AJ, Bhatti MS, Musharraf SG. Flow Injection-High Resolution-Electrospray Ionization-Mass Spectrometry (FI-HR-ESI-MS) Method for the Screening of Antimicrobial Pharmaceutical Drugs and Compounds against Klebsiella pneumoniae. Eur J Pharm Sci 2021; 157:105633. [PMID: 33130071 DOI: 10.1016/j.ejps.2020.105633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 01/07/2023]
Abstract
The development of drug resistant microorganism is a global threat. Therefore, screening of more compounds for antimicrobial potentials is needed. Hence, a rapid method was developed for the screening of antimicrobial drugs and compounds against Klebsiella pneumoniae using Flow Injection Analysis-High Resolution-Mass Spectrometry. The method was optimized for bacterial culture time and concentration of drugs. IC50 values for the drugs were calculated from the percent intensity of 704.5207 m/z of K. pneumoniae at 5 hrs incubation. This mass was proposed as diacylglycerophosphoethanolamine and observed as a potential biomarker of K. pneumoniae for the evaluation of inhibition potential of antimicrobial drugs and compounds. The calculated values for half maximal inhibitory concentration of cefixime, gentamicin and enrofloxacin were 0.052, 0.028 and 0.042 µg/mL, respectively. Ten compounds were also screened against the developed method, among them one compound (RSE-6) was found to be active with IC50 value of 45.08 µg/mL. The obtained results were further compared with MIC values, obtained from micro dilution and Alamar blue assay after 24 hrs incubation. In comparison to these methods, developed method is sensitive, reproducible, rapid and robust for the determination of IC50 value or inhibition potential of the drugs and compounds even at early incubation period of 5 hours.
Collapse
Affiliation(s)
- Muhammad Ramzan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Ayaz Ahmed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Yamina Usmani
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Amna Jabbar Siddiqui
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Salman Bhatti
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Syed Ghulam Musharraf
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
32
|
Peng Y, Cheng Z, Xie F. Evaluation of Pharmacokinetic Drug-Drug Interactions: A Review of the Mechanisms, In Vitro and In Silico Approaches. Metabolites 2021; 11:metabo11020075. [PMID: 33513941 PMCID: PMC7912632 DOI: 10.3390/metabo11020075] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/27/2022] Open
Abstract
Pharmacokinetic drug–drug interactions (DDIs) occur when a drug alters the absorption, transport, distribution, metabolism or excretion of a co-administered agent. The occurrence of pharmacokinetic DDIs may result in the increase or the decrease of drug concentrations, which can significantly affect the drug efficacy and safety in patients. Enzyme-mediated DDIs are of primary concern, while the transporter-mediated DDIs are less understood but also important. In this review, we presented an overview of the different mechanisms leading to DDIs, the in vitro experimental tools for capturing the factors affecting DDIs, and in silico methods for quantitative predictions of DDIs. We also emphasized the power and strategy of physiologically based pharmacokinetic (PBPK) models for the assessment of DDIs, which can integrate relevant in vitro data to simulate potential drug interaction in vivo. Lastly, we pointed out the future directions and challenges for the evaluation of pharmacokinetic DDIs.
Collapse
Affiliation(s)
| | | | - Feifan Xie
- Correspondence: ; Tel.: +86-0731-8265-0446
| |
Collapse
|
33
|
Kayesh R, Farasyn T, Crowe A, Liu Q, Pahwa S, Alam K, Neuhoff S, Hatley O, Ding K, Yue W. Assessing OATP1B1- and OATP1B3-Mediated Drug-Drug Interaction Potential of Vemurafenib Using R-Value and Physiologically-Based Pharmacokinetic Models. J Pharm Sci 2021; 110:314-324. [PMID: 32590030 PMCID: PMC7750294 DOI: 10.1016/j.xphs.2020.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 11/19/2022]
Abstract
Organic anion transporting polypeptides (OATP) 1B1 and OATP1B3 are important determinants of transporter-mediated drug-drug interactions (DDIs). Current studies assessed the OATP1B1 and OATP1B3-mediated DDI potential of vemurafenib, a kinase inhibitor drug with high protein binding and low aqueous solubility, using R-value and physiologically-based pharmacokinetic (PBPK) models. The total half-maximal inhibitory concentration (IC50,total) values of vemurafenib against OATP1B1 and OATP1B3 were determined in 100% human plasma in transporter-overexpressing human embryonic kidney 293 stable cell lines. The unbound fraction of vemurafenib in human plasma before (fu,plasma) and after addition into the uptake assay plate (fu,plasma,inc) were determined by rapid equilibrium dialysis. There was no statistically significant difference between fu,plasma and fu,plasma,inc. Vemurafenib IC50,total values against OATP1B1 and OATP1B3 are 175 ± 82 and 231 ± 26 μM, respectively. The R-values [R = 1 + fu,plasma × Iin,max/(fu,plasma,inc × IC50,total)] were then simplified as R = 1+Iin,max/IC50,total, and were 1.76 and 1.57 for OATP1B1 and OATP1B3, respectively. The simulated pravastatin AUC ratio was 1.28 when a single dose of pravastatin (40 mg) was co-administered with vemurafenib (960 mg, twice daily) at steady-state, compared to pravastatin alone. Both R-value and PBPK models predict that vemurafenib has the potential to cause OATP1B1- and OATP1B3-mediated DDIs.
Collapse
Affiliation(s)
- Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Taleah Farasyn
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Alexandra Crowe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Qiang Liu
- ARL Bio Pharma, Oklahoma City, Oklahoma 73104
| | - Sonia Pahwa
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Khondoker Alam
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Sibylle Neuhoff
- Certara UK Ltd, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ United Kingdom
| | - Oliver Hatley
- Certara UK Ltd, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ United Kingdom
| | - Kai Ding
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104.
| |
Collapse
|
34
|
Michiba K, Maeda K, Kurimori K, Enomoto T, Shimomura O, Takeuchi T, Nishiyama H, Oda T, Kusuhara H. Characterization of the Human Intestinal Drug Transport with Ussing Chamber System Incorporating Freshly Isolated Human Jejunum. Drug Metab Dispos 2021; 49:84-93. [PMID: 33087448 DOI: 10.1124/dmd.120.000138] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/02/2020] [Indexed: 02/13/2025] Open
Abstract
Intestinal permeability is a critical factor for orally administered drugs. It can be facilitated by uptake transporters or limited by efflux transporters and metabolic enzymes in the intestine. The present study aimed to characterize the Ussing chamber system incorporating human intestinal tissue as an in vitro model for investigating the impact of intestinal uptake/efflux transporters on the intestinal absorption of substrate drugs in humans. We confirmed the functions of major intestinal uptake/efflux drug transporters in freshly isolated human jejunum sections by demonstrating a significant decrease in the mucosal uptake of cefadroxil (peptide transporter 1) and methotrexate (proton-coupled folate transporter), mucosal-to-serosal permeability of ribavirin (concentrative nucleoside transporters/equilibrative nucleoside transporters), and serosal-to-mucosal permeability of P-glycoprotein and breast cancer resistance protein substrates in the presence of their typical inhibitors. The mucosal-to-serosal apparent permeability coefficients (Papp) of 19 drugs, including substrates of drug transporters and cytochrome P450 3A, ranged from 0.60 × 10-6 to 29 × 10-6 cm/s and showed a good correlation with reported fraction of an oral dose that enters the gut wall and passes into the portal circulation with escaping intestinal metabolism (FaFg) values in humans. Furthermore, the Papp values for cefadroxil, methotrexate, and ribavirin in the presence of the corresponding transporter inhibitors underestimated the FaFg of these drugs, which clearly showed that intestinal uptake transporters facilitate their intestinal absorption in humans. In conclusion, the functions of major intestinal uptake/efflux drug transporters could be maintained in freshly isolated human jejunum sections. The Ussing chamber system incorporating human intestinal tissue would be useful for evaluating the impact of intestinal uptake/efflux transporters on the intestinal absorption of various types of drugs in humans. SIGNIFICANCE STATEMENT: Although previous studies have predicted the intestinal absorption of drugs in humans using the Ussing chamber system incorporating human intestinal tissue, there is little systematic information about drug transport mediated by multiple transporters in this system. We confirmed the functions of major intestinal uptake/efflux transporters in freshly isolated human jejunum sections and demonstrated that the mucosal-to-serosal apparent permeability coefficient of various types of drugs showed a good correlation with reported human FaFg values.
Collapse
Affiliation(s)
- Kazuyoshi Michiba
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Ko Kurimori
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Tsuyoshi Enomoto
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Osamu Shimomura
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Tomoyo Takeuchi
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Hiroyuki Nishiyama
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Tatsuya Oda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| |
Collapse
|
35
|
Seoane RG, Garcia-Recio V, Garrosa M, Rojo MÁ, Jiménez P, Girbés T, Cordoba-Diaz M, Cordoba-Diaz D. Human Health Effects of Lactose Consumption as a Food and Drug Ingredient. Curr Pharm Des 2020; 26:1778-1789. [PMID: 32048961 DOI: 10.2174/1381612826666200212114843] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/01/2020] [Indexed: 02/07/2023]
Abstract
Lactose is a reducing sugar consisting of galactose and glucose, linked by a β (1→4) glycosidic bond, considered as an antioxidant due to its α-hydroxycarbonyl group. Lactose is widely ingested through the milk and other unfermented dairy products and is considered to be one of the primary foods. On the other hand, lactose is also considered as one of the most widely used excipients for the development of pharmaceutical formulations. In this sense, lactose has been related to numerous drug-excipient or drug-food pharmacokinetic interactions. Intolerance, maldigestion and malabsorption of carbohydrates are common disorders in clinical practice, with lactose-intolerance being the most frequently diagnosed, afflicting 10% of the world's population. Four clinical subtypes of lactose intolerance may be distinguished, namely lactase deficiency in premature infants, congenital lactase deficiency, adult-type hypolactasia and secondary lactase intolerance. An overview of the main uses of lactose in human nutrition and in the pharmaceutical industry and the problems derived from this circumstance are described in this review.
Collapse
Affiliation(s)
- Rafael G Seoane
- Area of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Verónica Garcia-Recio
- Area of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Manuel Garrosa
- Area of Histology, Faculty of Medicine and INCYL, University of Valladolid, 47005 Valladolid, Spain
| | - María Á Rojo
- Area of Experimental Sciences, Miguel de Cervantes European University, 47012 Valladolid, Spain
| | - Pilar Jiménez
- Area of Nutrition and Food Sciences, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - Tomás Girbés
- Area of Nutrition and Food Sciences, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - Manuel Cordoba-Diaz
- Area of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain.,University Institute of Industrial Pharmacy (IUFI), Complutense University of Madrid, 28040 Madrid, Spain
| | - Damián Cordoba-Diaz
- Area of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain.,University Institute of Industrial Pharmacy (IUFI), Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
36
|
Abstract
Amiodarone is an effective antiarrhythmic medication frequently used in practice for both ventricular and atrial arrhythmias. Though classified as a class III antiarrhythmic, it affects all phases of the cardiac action potential. However, the drug has several side effects, including thyroid abnormalities, pulmonary fibrosis, and transaminitis, for which routine monitoring is recommended. It also interacts with several medications, such as warfarin, simvastatin, and atorvastatin, and many HIV antiretroviral medications. Given the common use of this medication in medical practice, it is vital that clinicians understand the indications, contraindications, dosing, side effects, and interactions of this medication. A thorough understanding of these topics is essential for clinicians to ensure safe and effective use of amiodarone.
Collapse
|
37
|
Hakkola J, Hukkanen J, Turpeinen M, Pelkonen O. Inhibition and induction of CYP enzymes in humans: an update. Arch Toxicol 2020; 94:3671-3722. [PMID: 33111191 PMCID: PMC7603454 DOI: 10.1007/s00204-020-02936-7] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022]
Abstract
The cytochrome P450 (CYP) enzyme family is the most important enzyme system catalyzing the phase 1 metabolism of pharmaceuticals and other xenobiotics such as herbal remedies and toxic compounds in the environment. The inhibition and induction of CYPs are major mechanisms causing pharmacokinetic drug–drug interactions. This review presents a comprehensive update on the inhibitors and inducers of the specific CYP enzymes in humans. The focus is on the more recent human in vitro and in vivo findings since the publication of our previous review on this topic in 2008. In addition to the general presentation of inhibitory drugs and inducers of human CYP enzymes by drugs, herbal remedies, and toxic compounds, an in-depth view on tyrosine-kinase inhibitors and antiretroviral HIV medications as victims and perpetrators of drug–drug interactions is provided as examples of the current trends in the field. Also, a concise overview of the mechanisms of CYP induction is presented to aid the understanding of the induction phenomena.
Collapse
Affiliation(s)
- Jukka Hakkola
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, POB 5000, 90014, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Janne Hukkanen
- Biocenter Oulu, University of Oulu, Oulu, Finland.,Research Unit of Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Miia Turpeinen
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, POB 5000, 90014, Oulu, Finland.,Administration Center, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Olavi Pelkonen
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, POB 5000, 90014, Oulu, Finland.
| |
Collapse
|
38
|
Yulin Z, Lingti K, Shan G, Yong Z. A possible interaction between linezolid and digoxin: A case report of therapeutic drug monitoring. Saudi Pharm J 2020; 28:1408-1410. [PMID: 33250647 PMCID: PMC7679430 DOI: 10.1016/j.jsps.2020.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 09/08/2020] [Indexed: 11/06/2022] Open
Abstract
Drug-drug interactions lead to altered clinical effects, including adverse reactions. Therapeutic drug monitoring of digoxin is necessary due to its narrow therapeutic range. Linezolid can cause variable exposures in patients hospitalized in the intensive care unit owing to its possibility of drug-drug interactions. We present a patient with pneumonia and heart failure who experienced a possible drug interaction between linezolid and digoxin, resulting in high serum concentrations of both drugs. Also, the patient developed thrombocytopenia likely related to linezolid. The linezolid dose required to maintain sufficient levels had to reduce to 50% of the usual linezolid dose. A quarter dose of the standard digoxin dose was needed. Although the underlying mechanism of the drug interaction is unclear, we recommend conducting therapeutic drug monitoring when linezolid and digoxin are administered concurrently.
Collapse
Affiliation(s)
- Zhu Yulin
- Department of Pharmacy, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.,Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Kong Lingti
- Department of Pharmacy, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Gao Shan
- Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Zhang Yong
- Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
39
|
Arnold YE, Kalia YN. Using Ex Vivo Porcine Jejunum to Identify Membrane Transporter Substrates: A Screening Tool for Early-Stage Drug Development. Biomedicines 2020; 8:biomedicines8090340. [PMID: 32927779 PMCID: PMC7555276 DOI: 10.3390/biomedicines8090340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022] Open
Abstract
Robust, predictive ex vivo/in vitro models to study intestinal drug absorption by passive and active transport mechanisms are scarce. Membrane transporters can significantly impact drug uptake and transporter-mediated drug–drug interactions can play a pivotal role in determining the drug safety profile. Here, the presence and activity of seven clinically relevant apical/basolateral drug transporters found in human jejunum were tested using ex vivo porcine intestine in a Ussing chamber system. Experiments using known substrates of peptide transporter 1 (PEPT1), organic anion transporting polypeptide (OATP2B1), organic cation transporter 1 (OCT1), P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), multi drug resistance-associated protein 2 and 3 (MRP2 and MRP3), in the absence and presence of potent inhibitors, showed that there was a statistically significant change in apparent intestinal permeability Papp,pig (cm/s) in the presence of the corresponding inhibitor. For MRP2, a transporter reportedly present at relatively low concentration, although Papp,pig did not significantly change in the presence of the inhibitor, substrate deposition (QDEP) in the intestinal tissue was significantly increased. The activity of the seven transport proteins was successfully demonstrated and the results provided insight into their apical/basolateral localization. In conclusion, the results suggest that studies using the porcine intestine/Ussing chamber system, which could easily be integrated into the drug development process, might enable the early-stage identification of new molecular entities that are substrates of membrane transporters.
Collapse
Affiliation(s)
- Yvonne E. Arnold
- School of Pharmaceutical Sciences, University of Geneva, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland;
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Yogeshvar N. Kalia
- School of Pharmaceutical Sciences, University of Geneva, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland;
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland
- Correspondence: ; Tel.: +41-(0)22-379-3355
| |
Collapse
|
40
|
Impact of SLCO4C1 Genotypes, Creatinine, and Spironolactone on Digoxin Population Pharmacokinetic Variables in Patients With Cardiac Insufficiency. Clin Ther 2020; 42:1799-1810.e3. [DOI: 10.1016/j.clinthera.2020.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/02/2020] [Accepted: 07/17/2020] [Indexed: 01/18/2023]
|
41
|
Ashrafizadeh M, Zarrabi A, Orouei S, Saberifar S, Salami S, Hushmandi K, Najafi M. Recent advances and future directions in anti-tumor activity of cryptotanshinone: A mechanistic review. Phytother Res 2020; 35:155-179. [PMID: 33507609 DOI: 10.1002/ptr.6815] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/29/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
In respect to the enhanced incidence rate of cancer worldwide, studies have focused on cancer therapy using novel strategies. Chemotherapy is a common strategy in cancer therapy, but its adverse effects and chemoresistance have limited its efficacy. So, attempts have been directed towards minimally invasive cancer therapy using plant derived-natural compounds. Cryptotanshinone (CT) is a component of salvia miltiorrihiza Bunge, well-known as Danshen and has a variety of therapeutic and biological activities such as antioxidant, anti-inflammatory, anti-diabetic and neuroprotective. Recently, studies have focused on anti-tumor activity of CT against different cancers. Notably, this herbal compound is efficient in cancer therapy by targeting various molecular signaling pathways. In the present review, we mechanistically describe the anti-tumor activity of CT with an emphasis on molecular signaling pathways. Then, we evaluate the potential of CT in cancer immunotherapy and enhancing the efficacy of chemotherapy by sensitizing cancer cells into anti-tumor activity of chemotherapeutic agents, and elevating accumulation of anti-tumor drugs in cancer cells. Finally, we mention strategies to enhance the anti-tumor activity of CT, for instance, using nanoparticles to provide targeted drug delivery.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey.,Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul, Turkey
| | - Sima Orouei
- MSc. Student, Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sedigheh Saberifar
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Saeed Salami
- DVM. Graduated, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
42
|
Deng F, Ghemtio L, Grazhdankin E, Wipf P, Xhaard H, Kidron H. Binding Site Interactions of Modulators of Breast Cancer Resistance Protein, Multidrug Resistance-Associated Protein 2, and P-Glycoprotein Activity. Mol Pharm 2020; 17:2398-2410. [PMID: 32496785 PMCID: PMC7497665 DOI: 10.1021/acs.molpharmaceut.0c00155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
ATP-binding cassette (ABC)-transporters
protect tissues by pumping
their substrates out of the cells in many physiological barriers,
such as the blood–brain barrier, intestine, liver, and kidney.
These substrates include various endogenous metabolites, but, in addition,
ABC transporters recognize a wide range of compounds, therefore affecting
the disposition and elimination of clinically used drugs and their
metabolites. Although numerous ABC-transporter inhibitors are known,
the underlying mechanism of inhibition is not well characterized.
The aim of this study is to deepen our understanding of transporter
inhibition by studying the molecular basis of ligand recognition.
In the current work, we compared the effect of 44 compounds on the
active transport mediated by three ABC transporters: breast cancer
resistance protein (BCRP and ABCG2), multidrug-resistance associated
protein (MRP2 and ABCC2), and P-glycoprotein (P-gp and ABCB1). Eight
compounds were strong inhibitors of all three transporters, while
the activity of 36 compounds was transporter-specific. Of the tested
compounds, 39, 25, and 11 were considered as strong inhibitors, while
1, 4, and 11 compounds were inactive against BCRP, MRP2, and P-gp,
respectively. In addition, six transport-enhancing stimulators were
observed for P-gp. In order to understand the observed selectivity,
we compared the surface properties of binding cavities in the transporters
and performed structure–activity analysis and computational
docking of the compounds to known binding sites in the transmembrane
domains and nucleotide-binding domains. Based on the results, the
studied compounds are more likely to interact with the transmembrane
domain than the nucleotide-binding domain. Additionally, the surface
properties of the substrate binding site in the transmembrane domains
of the three transporters were in line with the observed selectivity.
Because of the high activity toward BCRP, we lacked the dynamic range
needed to draw conclusions on favorable interactions; however, we
identified amino acids in both P-gp and MRP2 that appear to be important
for ligand recognition.
Collapse
Affiliation(s)
- Feng Deng
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Leo Ghemtio
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Evgeni Grazhdankin
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Peter Wipf
- Department of Chemistry, The Center for Chemical Methodologies and Library Development, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Henri Xhaard
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki 00014, Finland
| |
Collapse
|
43
|
Negoro R, Kawai K, Ichikawa M, Deguchi S, Takayama K, Mizuguchi H. Establishment of MDR1-knockout human induced pluripotent stem cell line. Drug Metab Pharmacokinet 2020; 35:288-296. [PMID: 32303458 DOI: 10.1016/j.dmpk.2020.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022]
Abstract
Multiple drug resistance 1 (MDR1) is highly expressed in various organs, including the liver, small intestine, and blood-brain barrier (BBB). Because MDR1 plays important roles in the excretion of many drugs, it is necessary to evaluate whether drug candidates are potential substrates of MDR1. Recently, many researchers have shown that human induced pluripotent stem (iPS) cell-derived differentiated cells such as hepatocytes and enterocytes can be applied for pharmacokinetic testing. Here, we attempted to generate MDR1-knockout (KO) iPS cell lines using genome editing technology. The correctly targeted human iPS cell lines were successfully obtained. The expression levels of pluripotent markers in human iPS cells were not changed by MDR1 knockout. The gene expression levels of hepatic markers in MDR1-KO iPS-derived hepatocyte-like cells were higher than those in undifferentiated MDR1-KO iPS cells, suggesting that MDR1-KO iPS cells have hepatic differentiation capacity. In addition, MDR1 expression levels were hardly detected in MDR1-KO iPS cell-derived hepatocyte-like cells. We thus succeeded in establishing MDR1-KO iPS cell lines that could be utilized for pharmacokinetic testing.
Collapse
Affiliation(s)
- Ryosuke Negoro
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Kanae Kawai
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Moe Ichikawa
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Sayaka Deguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan
| | - Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; PRESTO, Japan Science and Technology Agency, Saitama, 332-0012, Japan.
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
44
|
Thibault N, Ibrahim J, Peterschmitt MJ, Puga AC, Ross L, Vu L, Xue Y, Turpault S. Effect of eliglustat on the pharmacokinetics of digoxin, metoprolol, and oral contraceptives and absorption of eliglustat when coadministered with acid-reducing agents. Mol Genet Metab 2020; 129:278-285. [PMID: 32029355 DOI: 10.1016/j.ymgme.2020.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/25/2019] [Accepted: 01/06/2020] [Indexed: 11/30/2022]
Abstract
Eliglustat is an oral substrate reduction therapy indicated for patients with Gaucher disease type 1. Based on in vitro data, clinical trials were conducted to assess the potential for drug-drug interactions between eliglustat and digoxin (P-glycoprotein substrate), metoprolol (sensitive CYP2D6 substrate), a combined oral contraceptive (CYP3A substrate), and acid-reducing agents. Healthy subjects were enrolled in four Phase 1 clinical studies to evaluate the effect of eliglustat on the pharmacokinetics, safety, and tolerability of digoxin (N = 28), metoprolol (N = 14), and a combined oral contraceptive (N = 30) and the effect of acid-reducing agents on eliglustat pharmacokinetics, safety, and tolerability (N = 24). Coadministration resulted in increased exposure to digoxin (1.49-fold) and metoprolol (2-fold) with eliglustat, negligible effects on oral contraceptive pharmacokinetics with eliglustat, and a negligible effect of acid-reducing agents on eliglustat pharmacokinetics. Across all studies, eliglustat was well-tolerated. One serious adverse event (spontaneous abortion) and one discontinuation due to an adverse event (urinary tract infection) were reported, both during the acid-reducing agents study. When eliglustat is coadministered with medications that are P-glycoprotein or CYP2D6 substrates, lower doses of these concomitant medications may be required. Eliglustat may be coadministered with oral contraceptives and acid-reducing agents without dose modifications for either drug.
Collapse
Affiliation(s)
| | | | | | | | | | - Lucie Vu
- Sanofi Genzyme, Cambridge, MA, USA
| | - Yong Xue
- Sanofi Genzyme, Cambridge, MA, USA
| | | |
Collapse
|
45
|
Value of quantifying ABC transporters by mass spectrometry and impact on in vitro-to-in vivo prediction of transporter-mediated drug-drug interactions of rivaroxaban. Eur J Pharm Biopharm 2020; 148:27-37. [PMID: 31945490 DOI: 10.1016/j.ejpb.2020.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/04/2019] [Accepted: 01/10/2020] [Indexed: 11/20/2022]
Abstract
ABC transporters, such as P-gp and BCRP, are involved in rivaroxaban pharmacokinetics and can lead to drug-drug interactions (DDIs). Investigations of the victim role for rivaroxaban and transporter-mediated DDI are commonly performed using in vitro models. However, interpretation of rivaroxaban efflux transport and DDI studies in cell models may be influenced by P-gp and BCRP transporter abundance. This study aimed to develop an LC-MS/MS quantification method for assessing the relationship between transporter expression and functionality in Caco-2ATCC, Caco-2ECACC, MDCK-MDR1, MDCK-BCRP cell models. First, the relative and absolute quantities of the transporters were determined by LC-MS/MS. P-gp and BCRP expression was then confirmed by western blotting and immunofluorescence staining. Finally, P-gp and BCRP functional activities and half-inhibitory concentrations (IC50s) of two specific inhibitors (verapamil and ko143) were determined by bidirectional transport experiments. P-gp and BCRP protein expression was detected at the cell membrane and was greater in the respective transfected models. Efflux ratios were correlated with P-gp and BCRP quantities. The lowest IC50s were obtained in the MDCK-MDR1 and MDCK-BCRP models for verapamil and ko143, respectively. In conclusion, this study demonstrated that LC-MS/MS can accurately quantify P-gp and BCRP efflux transporters and thereby improve the interpretation of transport data and in vitro-in vivo correlations.
Collapse
|
46
|
Role of P-glycoprotein in the brain disposition of seletalisib: Evaluation of the potential for drug-drug interactions. Eur J Pharm Sci 2020; 142:105122. [PMID: 31678424 DOI: 10.1016/j.ejps.2019.105122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 11/21/2022]
Abstract
Seletalisib is an orally bioavailable selective inhibitor of phosphoinositide 3-kinase delta (PI3Kδ) in clinical development for the treatment of immune-mediated inflammatory diseases. The present study investigated the role of P-gp in seletalisib disposition, especially brain distribution, and the associated risks of interactions. Seletalisib was found to be actively transported by rodent and human P-gp in vitro (transfected LLC-PK1 cells; Km of ca. 20 µM), with minimal or no affinity for the other tested transporters. A distribution study in knockout rats (single oral dosing at 750 mg kg-1) showed that P-gp restricts the brain disposition of seletalisib while having minimal effect on its intestinal absorption. Restricted brain penetration was also observed in cynomolgus monkeys (single oral dosing at 30 mg kg-1) using brain microdialysis and cerebrospinal fluid sampling (Kp,uu of 0.09 and 0.24, respectively). These findings opened the question of potential pharmacokinetic interaction between seletalisib and P-gp inhibitors. In vitro, CsA inhibited the active transport of seletalisib with an IC50 of 0.13 µM. In rats, co-administration of high doses of CsA (bolus iv followed by continuous infusion) increased the brain distribution of seletalisib (single oral dosing at 5 mg kg-1). The observed data were found aligned with those predicted by in vitro-in vivo extrapolation. Based on the same extrapolation method combined with literature data, only very few P-gp inhibitors (i.e. CsA, quinine, quinidine) were predicted to increase the brain disposition of seletalisib in the clinical setting (maximal 3-fold changes).
Collapse
|
47
|
Aubets J, Jansat J, Salva M, Birks VM, Cole RJ, Lewis J, Pitcher A, Hall M. No evidence for interactions of dimethylfumarate (DMF) and its main metabolite monomethylfumarate (MMF) with human cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) drug transporter. Pharmacol Res Perspect 2019; 7:e00540. [PMID: 31832203 PMCID: PMC6887663 DOI: 10.1002/prp2.540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 01/28/2023] Open
Abstract
Dimethylfumarate (DMF) has long been used as part of a fixed combination of fumaric acid esters (FAE) in some European countries and is now available as an oral monotherapy for psoriasis. The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF's regulatory commitments. Although referred to in the available product labels/summary of product characteristics, the actual data have not yet been made publicly available. In vitro inhibition experiments using CYP-selective substrates with human liver microsomes showed 50% inhibitory concentrations (IC50) of >666 µmol/L for DMF and >750 µmol/L for MMF. MMF (≤250 μmol/L; 72 hours) was not cytotoxic in cultured human hepatocyte experiments and mRNA expression data indicated no CYP induction by MMF (1-250 µmol/L). DMF (≤6.66 mmol/L) showed moderate-to-high absorption (apparent permeability [Papp] ≥2.3-29.7 x 10-6 cm/s) across a Caucasian colon adenocarcinoma (Caco-2) cell monolayer, while MMF (≤7.38 mmol/L) demonstrated low-to-moderate permeability (Papp 1.2-8.9 × 10-6 cm/s). DMF was not a substrate for P-gp (net efflux ratios ≤1.22) but was a weak inhibitor of P-gp at supratherapeutic concentrations (estimated IC50 relative to solvent control of 1.5 mmol/L; [3H]digoxin efflux in Caco-2 cells). This inhibition is unlikely to be clinically relevant. MMF was not a substrate or inhibitor of P-gp. Thus, DMF and MMF should not affect the absorption, distribution, metabolism or excretion of coadministered drugs that are CYP and P-gp substrates.
Collapse
Affiliation(s)
- Jordi Aubets
- Department of DMPK DevelopmentAlmirall S.A.BarcelonaSpain
| | | | - Miquel Salva
- Department of DMPK DevelopmentAlmirall S.A.BarcelonaSpain
| | | | | | | | | | | |
Collapse
|
48
|
Joshi PS, Sanakkayala N, Kirkpatrick L, Terse PS. Nonclinical safety assessment of PF614: A novel TAAP prodrug of oxycodone for chronic pain indication. Regul Toxicol Pharmacol 2019; 108:104433. [PMID: 31362032 PMCID: PMC6790176 DOI: 10.1016/j.yrtph.2019.104433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/03/2019] [Accepted: 07/26/2019] [Indexed: 11/26/2022]
Abstract
PF614, a novel trypsin activated abuse protection (TAAP) prodrug of oxycodone, is being studied as chronic pain analgesic with extended release and abuse resistant properties. A series of nonclinical safety studies were conducted to support PF614 introduction to clinical trials. Ames assays (PF614 and its metabolites), comet assay (PF614 ≤ 50 mg/kg/day oral gavage in rats) and micronucleus assay (PF614 ≤ 175 mg/kg/day oral gavage in rats) were negative. hERG assay IC50 for PF614 was ≥300 μM. PF614 (0.1 and 10 μM) showed a low permeability in Caco-2 cells (≤1.17 x 10-6 cm/s) and was not a P-gp or BCRP substrate or inhibitor. The mean percent unbound PF614 among all concentrations in plasma ranged from 91.2 to 98.4, 79.4 to 100, and 52.9-79.9% in rat, dog, and human, respectively. Also, PF614 was metabolically stable in rat, dog, and human hepatocytes with no metabolites identified. Safety pharmacology study in dog indicated moderately lower heart rate at ≥ 2 mg/kg oral gavage doses. Toxicity studies of PF614 in rat and dog with daily oral doses of 25 and 18 mg/kg, respectively, for 14 Days were well tolerated with favorable safety profile supporting its further clinical evaluation.
Collapse
Affiliation(s)
- P S Joshi
- National Center for Advancing Translational Sciences, Bethesda, MD, United States
| | - N Sanakkayala
- Covance Laboratories, Inc., 3301 Kinsman Blvd., Madison, WI, United States
| | - L Kirkpatrick
- Ensysce Biosciences Inc., San Diego, CA, United States
| | - P S Terse
- National Center for Advancing Translational Sciences, Bethesda, MD, United States.
| |
Collapse
|
49
|
Yamazaki S, Costales C, Lazzaro S, Eatemadpour S, Kimoto E, Varma MV. Physiologically-Based Pharmacokinetic Modeling Approach to Predict Rifampin-Mediated Intestinal P-Glycoprotein Induction. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:634-642. [PMID: 31420942 PMCID: PMC6765699 DOI: 10.1002/psp4.12458] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/24/2019] [Indexed: 12/25/2022]
Abstract
Physiologically‐based pharmacokinetic (PBPK) modeling is a powerful tool to quantitatively describe drug disposition profiles in vivo, thereby providing an alternative to predict drug–drug interactions (DDIs) that have not been tested clinically. This study aimed to predict effects of rifampin‐mediated intestinal P‐glycoprotein (Pgp) induction on pharmacokinetics of Pgp substrates via PBPK modeling. First, we selected four Pgp substrates (digoxin, talinolol, quinidine, and dabigatran etexilate) to derive in vitro to in vivo scaling factors for intestinal Pgp kinetics. Assuming unbound Michaelis‐Menten constant (Km) to be intrinsic, we focused on the scaling factors for maximal efflux rate (Jmax) to adequately recover clinically observed results. Next, we predicted rifampin‐mediated fold increases in intestinal Pgp abundances to reasonably recover clinically observed DDI results. The modeling results suggested that threefold to fourfold increases in intestinal Pgp abundances could sufficiently reproduce the DDI results of these Pgp substrates with rifampin. Hence, the obtained fold increases can potentially be applicable to DDI prediction with other Pgp substrates.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, San Diego, California, USA
| | - Chester Costales
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Groton, Connecticut, USA
| | - Sarah Lazzaro
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Groton, Connecticut, USA
| | - Soraya Eatemadpour
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Groton, Connecticut, USA
| | - Emi Kimoto
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Groton, Connecticut, USA
| | - Manthena V Varma
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, Groton, Connecticut, USA
| |
Collapse
|
50
|
Angeles PC, Robertsen I, Seeberg LT, Krogstad V, Skattebu J, Sandbu R, Åsberg A, Hjelmesæth J. The influence of bariatric surgery on oral drug bioavailability in patients with obesity: A systematic review. Obes Rev 2019; 20:1299-1311. [PMID: 31232513 PMCID: PMC6852510 DOI: 10.1111/obr.12869] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/30/2019] [Accepted: 03/31/2019] [Indexed: 12/16/2022]
Abstract
Anatomical changes in the gastrointestinal tract and subsequent weight loss may influence drug disposition and thus drug dosing following bariatric surgery. This review systematically examines the effects of bariatric surgery on drug pharmacokinetics, focusing especially on the mechanisms involved in restricting oral bioavailability. Studies with a longitudinal before-after design investigating the pharmacokinetics of at least one drug were reviewed. The need for dose adjustment following bariatric surgery was examined, as well as the potential for extrapolation to other drugs subjected to coinciding pharmacokinetic mechanisms. A total of 22 original articles and 32 different drugs were assessed. The majority of available data is based on Roux-en-Y gastric bypass (RYGBP) (18 of 22 studies), and hence, the overall interpretation is more or less limited to RYGBP. In the case of the majority of studied drugs, an increased absorption rate was observed early after RYGBP. The effect on systemic exposure allows for a low degree of extrapolation, including between drugs subjected to the same major metabolic and transporter pathways. On the basis of current understanding, predicting the pharmacokinetic change for a specific drug following RYGBP is challenging. Close monitoring of each individual drug is therefore recommended in the early postsurgical phase. Future studies should focus on the long-term effects of bariatric surgery on drug disposition, and they should also aim to disentangle the effects of the surgery itself and the subsequent weight loss.
Collapse
Affiliation(s)
- Philip Carlo Angeles
- Morbid Obesity Centre, Department of MedicineVestfold Hospital TrustTønsbergNorway
- Department of SurgeryVestfold Hospital TrustTønsbergNorway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Ida Robertsen
- Section of Pharmacology and Pharmaceutical Biosciences, Department of PharmacyUniversity of OsloOsloNorway
| | | | - Veronica Krogstad
- Section of Pharmacology and Pharmaceutical Biosciences, Department of PharmacyUniversity of OsloOsloNorway
| | - Julie Skattebu
- Library of Health SciencesVestfold Hospital TrustTønsbergNorway
| | - Rune Sandbu
- Morbid Obesity Centre, Department of MedicineVestfold Hospital TrustTønsbergNorway
- Department of SurgeryVestfold Hospital TrustTønsbergNorway
| | - Anders Åsberg
- Section of Pharmacology and Pharmaceutical Biosciences, Department of PharmacyUniversity of OsloOsloNorway
- Department of Transplantation MedicineOslo University Hospital‐RikshospitaletOsloNorway
| | - Jøran Hjelmesæth
- Morbid Obesity Centre, Department of MedicineVestfold Hospital TrustTønsbergNorway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
| |
Collapse
|