1
|
Pan C, Lee LTO. Membrane drug transporters in cancer: From chemoresistance mechanism to therapeutic strategies. Biochim Biophys Acta Rev Cancer 2025; 1880:189272. [PMID: 39863184 DOI: 10.1016/j.bbcan.2025.189272] [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: 10/07/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Chemoresistance is a multifactorial phenomenon and the primary cause to the ineffectiveness of oncotherapy and cancer recurrence. Membrane drug transporters are crucial for drug delivery and disposition in cancer cells. Changes in the expression and functionality of these transporters lead to decreased intracellular accumulation and reduced toxicity of antineoplastic drugs. As the mechanism has been better understood and genetic engineering technology progressed quickly in recent years, some novel targeting strategies have come to light. This article summarizes the regulatory mechanisms of membrane drug transporters and provides an extensive review of current approaches to address transporters-mediated chemoresistance. These strategies include the use of chemical inhibitors to block efflux transporters, the development of copper chelators to enhance platinum drug uptake, the delivery of genetic drugs to alter transporter expression, the regulation of transcription and post-translational modifications. Additionally, we provide information of the clinical trial performance of the related targeting strategies, along with the ongoing challenges. Even though some clinical trials failed due to unexpected side effects and limited therapeutic efficacy, the advent of targeting membrane drug transporters still presents a hopeful path for overcoming chemoresistance.
Collapse
Affiliation(s)
- Chao Pan
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Leo Tsz On Lee
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China.
| |
Collapse
|
2
|
Dermota T, Jug B, Trontelj J, Božič Mijovski M. Ticagrelor is Associated with Increased Rosuvastatin Blood Concentrations in Patients who have had a Myocardial Infarction. Clin Pharmacokinet 2025; 64:565-571. [PMID: 40029501 PMCID: PMC12041183 DOI: 10.1007/s40262-025-01489-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 03/05/2025]
Abstract
AIMS The primary objective of this study was to determine whether concomitant therapy with ticagrelor and rosuvastatin affects rosuvastatin plasma concentrations in patients receiving rosuvastatin 40 mg/day after myocardial infarction. METHODS We included 93 patients who had experienced a myocardial infarction and were receiving high-dose rosuvastatin 40 mg/day and a P2Y12 receptor antagonist, either ticagrelor, prasugrel or clopidogrel. We used liquid chromatography with tandem mass spectrometry to measure rosuvastatin plasma concentrations after liquid-liquid extraction. RESULTS Rosuvastatin plasma concentrations (9.7 ng/mL) were approximately twice as high in patients receiving ticagrelor therapy as in those receiving prasugrel (5.1 ng/mL, p < 0.001) or clopidogrel (5.0 ng/mL, p = 0.009), and ticagrelor was an independent factor influencing rosuvastatin concentrations. In addition, creatinine levels were associated with increased rosuvastatin concentrations (p = 0.039). CONCLUSION Our results suggest an important pharmacokinetic interaction between ticagrelor and rosuvastatin, leading to approximately two-fold higher rosuvastatin plasma concentrations in those receiving concomitant ticagrelor than in those receiving prasugrel or clopidogrel. The association is significant and independent of other potential factors influencing rosuvastatin levels, indicating its potential clinical relevance.
Collapse
Affiliation(s)
- Tjaša Dermota
- University Medical Centre Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia.
| | - Borut Jug
- University Medical Centre Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - Jurij Trontelj
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
3
|
Schaller S, Michon I, Baier V, Martins FS, Nolain P, Taneja A. Evaluation of BCRP-Related DDIs Between Methotrexate and Cyclosporin A Using Physiologically Based Pharmacokinetic Modelling. Drugs R D 2025; 25:1-17. [PMID: 39715910 PMCID: PMC12011704 DOI: 10.1007/s40268-024-00495-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVE This study provides a physiologically based pharmacokinetic (PBPK) model-based analysis of the potential drug-drug interaction (DDI) between cyclosporin A (CsA), a breast cancer resistance protein transporter (BCRP) inhibitor, and methotrexate (MTX), a putative BCRP substrate. METHODS PBPK models for CsA and MTX were built using open-source tools and published data for both model building and for model verification and validation. The MTX and CsA PBPK models were evaluated for their application in simulating BCRP-related DDIs. A qualification of an introduced empirical uniform in vitro scaling factor of Ki values for transporter inhibition by CsA was conducted by using a previously developed model of rosuvastatin (sensitive index BCRP substrate), and assessing if corresponding DDI ratios were well captured. RESULTS Within the simulated DDI scenarios for MTX in the presence of CsA, the developed models could capture the observed changes in PK parameters as changes in the area under the curve ratios (area under the curve during DDI/area under the curve control) of 1.30 versus 1.31 observed and the DDI peak plasma concentration ratios (peak plasma concentration during DDI/peak plasma concentration control) of 1.07 versus 1.28 observed. The originally reported in vitro Ki values of CsA were scaled with the uniform qualified scaling factor for their use in the in vivo DDI simulations to correct for the low intracellular unbound fraction of the CsA effector concentration. The resulting predicted versus observed ratios of peak plasma concentration and area under the curve DDI ratios with MTX were 0.82 and 0.99, respectively, indicating adequate model accuracy and choice of a scaling factor to capture the observed DDI. CONCLUSIONS All models have been comprehensively documented and made publicly available as tools to support the drug development and clinical research community and further community-driven model development.
Collapse
Affiliation(s)
| | | | | | | | | | - Amit Taneja
- Galapagos SASU, Romainville, France
- Simulations Plus, Inc., Lancaster, California, USA
| |
Collapse
|
4
|
Huang L, Ning C, He J, Wang M, Chen X, Guo X, Zhong L. Evaluation of drug-drug interaction between rosuvastatin and tacrolimus and the risk of hepatic injury in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03768-3. [PMID: 39862262 DOI: 10.1007/s00210-024-03768-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025]
Abstract
Multimorbidity, therapeutic complexity, and polypharmacy, which greatly increases the risk of drug-drug interactions (DDIs) and adverse medical outcomes, have become important and growing challenges in clinical practice. Statins are frequently prescribed to manage post-transplant dyslipidemia and reduce overall cardiovascular risk in solid organ transplant recipients. This study aimed to determine whether rosuvastatin has significant DDIs with tacrolimus (the first-line immunosuppressant) and to evaluate the risk of hepatotoxicity associated with concomitant therapy. We first studied whether a rat model could be established to assess the magnitude of rosuvastatin-tacrolimus DDI. The liver function index and histopathological examination were performed to investigate the characteristics of hepatotoxicity in the presence and absence of DDI. The clinical DDI potential between rosuvastatin and tacrolimus was also explored. Single-dose intravenous administration of tacrolimus did not significantly affect the area under the plasma concentration-time curve (AUC0-∞), clearance (CL), and volume of distribution at steady-state (Vss) of rosuvastatin in rats, despite a 96.7% increase in the rosuvastatin maximum plasma concentration (P = 0.024). Multiple doses of intravenous tacrolimus had no effect on the systemic disposition of rosuvastatin, but significantly increased aspartate transaminase (AST) by 42.6% (P = 0.043). Multiple doses of intravenous tacrolimus and rosuvastatin significantly altered the disposition of rosuvastatin, reducing alanine aminotransferase (ALT) and AST by 38.3% (P = 0.040) and 31.6% (P = 0.019), respectively. Histological evaluation of the liver specimens revealed patterns of drug-induced liver injury in rats. At clinically relevant doses, tacrolimus was predicted to be unable to cause pharmacokinetic interactions with rosuvastatin through basic models. The concomitant administration of tacrolimus and rosuvastatin has a minor impact on rosuvastatin pharmacokinetics; however, mild hepatotoxicity has been observed in rats.
Collapse
Affiliation(s)
- Lulu Huang
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Bayi Avenue No. 461, Nanchang, 330006, Jiangxi, China
| | - Chen Ning
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Jiake He
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi, China.
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi, China.
- Department of Pharmacy, Nanchang County People's Hospital, Xiangyang Road No. 199, Nanchang, 330200, Jiangxi, China.
| | - Mingcheng Wang
- Department of Pharmacy, Nanchang County People's Hospital, Xiangyang Road No. 199, Nanchang, 330200, Jiangxi, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Xiaohui Guo
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi, China
| | - Lin Zhong
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
5
|
Kiełbowski K, Król M, Bakinowska E, Pawlik A. The Role of ABCB1, ABCG2, and SLC Transporters in Pharmacokinetic Parameters of Selected Drugs and Their Involvement in Drug-Drug Interactions. MEMBRANES 2024; 14:223. [PMID: 39590609 PMCID: PMC11596214 DOI: 10.3390/membranes14110223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024]
Abstract
Membrane transporters are expressed in a wide range of tissues in the human organism. These proteins regulate the penetration of various substances such as simple ions, xenobiotics, and an extensive number of therapeutics. ABC and SLC drug transporters play a crucial role in drug absorption, distribution, and elimination. Recent decades have shown their contribution to the systemic exposure and tissue penetration of numerous drugs, thereby having an impact on pharmacokinetic and pharmacodynamic parameters. Importantly, the activity and expression of these transporters depend on numerous conditions, including intestinal microbiome profiles or health conditions. Moreover, the combined intake of other drugs or natural agents further affects the functionality of these proteins. In this review, we will discuss the involvement of ABC and SLC transporters in drug disposition. Moreover, we will present current evidence of the potential role of drug transporters as therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (M.K.); (E.B.)
| |
Collapse
|
6
|
Shen H, Huo R, Zhang Y, Wang L, Tong N, Chen W, Paris AJ, Mensah K, Chen M, Xue Y, Li W, Sinz M. A Pilot Study To Assess the Suitability of Riboflavin As a Surrogate Marker of Breast Cancer Resistance Protein in Healthy Participants. J Pharmacol Exp Ther 2024; 390:162-173. [PMID: 38296646 DOI: 10.1124/jpet.123.002015] [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: 11/16/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
We recently showed that riboflavin is a selected substrate of breast cancer resistance protein (BCRP) over P-glycoprotein (P-gp) and demonstrated its prediction performance in preclinical drug-drug interaction (DDI) studies. The aim of this study was to investigate the suitability of riboflavin to assess BCRP inhibition in humans. First, we assessed the substrate potential of riboflavin toward other major drug transporters using established transfected cell systems. Riboflavin is a substrate for organic anion transporter (OAT)1, OAT3, and multidrug and toxin extrusion protein (MATE)2-K, with uptake ratios ranging from 2.69 to 11.6, but riboflavin is not a substrate of organic anion-transporting polypeptide (OATP)1B1, OATP1B3, organic cation transporter (OCT)2, and MATE1. The effects of BMS-986371, a potent in vitro inhibitor of BCRP (IC 50 0.40 μM), on the pharmacokinetics of riboflavin, isobutyryl carnitine, and arginine were then examined in healthy male adults (N = 14 or 16) after oral administration of methotrexate (MTX) (7.5 mg) and enteric-coated (EC) sulfasalazine (SSZ) (1000 mg) alone or in combination with BMS-986371 (150 mg). Oral administration of BMS-986371 increased the area under the plasma concentration-time curves (AUCs) of rosuvastatin and immediate-release (IR) SSZ to 1.38- and 1.51-fold, respectively, and significantly increased AUC(0-4h), AUC(0-24h), and C max of riboflavin by 1.25-, 1.14-, and 1.11-fold (P-values of 0.003, 0.009, and 0.025, respectively) compared with the MTX/SSZ EC alone group. In contrast, BMS-986371 did not significantly influence the AUC(0-24h) and C max values of isobutyryl carnitine and arginine (0.96- to 1.07-fold, respectively; P > 0.05). Overall, these data indicate that plasma riboflavin is a promising biomarker of BCRP that may offer a possibility to assess drug candidate as a BCRP modulator in early drug development. SIGNIFICANCE STATEMENT: Endogenous compounds that serve as biomarkers for clinical inhibition of breast cancer resistance protein (BCRP) are not currently available. This study provides the initial evidence that riboflavin is a promising BCRP biomarker in humans. For the first time, the value of leveraging the substrate of BCRP with acceptable prediction performance in clinical studies is shown. Additional clinical investigations with known BCRP inhibitors are needed to fully validate and showcase the utility of this biomarker.
Collapse
Affiliation(s)
- Hong Shen
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Runlan Huo
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Yueping Zhang
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Linna Wang
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Nian Tong
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Weiqi Chen
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Andrew J Paris
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Kofi Mensah
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Min Chen
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Yongjun Xue
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Wenying Li
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| | - Michael Sinz
- Departments of Drug Metabolism and Pharmacokinetics (H.S., Y.Z., M.S.), Clinical Pharmacology, Pharmacometrics, and Bioanalysis (R.H., L.W., M.C., Y.X.), Development Biotransformation (N.T., W.C., W.L.), and Early Clinical Development (A.J.P., K.M.), Bristol Myers Squibb, Princeton, New Jersey
| |
Collapse
|
7
|
Russell LE, Yadav J, Maldonato BJ, Chien HC, Zou L, Vergara AG, Villavicencio EG. Transporter-mediated drug-drug interactions: regulatory guidelines, in vitro and in vivo methodologies and translation, special populations, and the blood-brain barrier. Drug Metab Rev 2024:1-28. [PMID: 38967415 DOI: 10.1080/03602532.2024.2364591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024]
Abstract
This review, part of a special issue on drug-drug interactions (DDIs) spearheaded by the International Society for the Study of Xenobiotics (ISSX) New Investigators, explores the critical role of drug transporters in absorption, disposition, and clearance in the context of DDIs. Over the past two decades, significant advances have been made in understanding the clinical relevance of these transporters. Current knowledge on key uptake and efflux transporters that affect drug disposition and development is summarized. Regulatory guidelines from the FDA, EMA, and PMDA that inform the evaluation of potential transporter-mediated DDIs are discussed in detail. Methodologies for preclinical and clinical testing to assess potential DDIs are reviewed, with an emphasis on the utility of physiologically based pharmacokinetic (PBPK) modeling. This includes the application of relative abundance and expression factors to predict human pharmacokinetics (PK) using preclinical data, integrating the latest regulatory guidelines. Considerations for assessing transporter-mediated DDIs in special populations, including pediatric, hepatic, and renal impairment groups, are provided. Additionally, the impact of transporters at the blood-brain barrier (BBB) on the disposition of CNS-related drugs is explored. Enhancing the understanding of drug transporters and their role in drug disposition and toxicity can improve efficacy and reduce adverse effects. Continued research is essential to bridge remaining gaps in knowledge, particularly in comparison with cytochrome P450 (CYP) enzymes.
Collapse
Affiliation(s)
- Laura E Russell
- Department of Quantitative, Translational, and ADME Sciences, AbbVie Inc, North Chicago, IL, USA
| | - Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Boston, MA, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc, Redwood City, CA, USA
| | - Huan-Chieh Chien
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ling Zou
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ana G Vergara
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Rahway, NJ, USA
| | - Erick G Villavicencio
- Department of Biology-Discovery, Imaging and Functional Genomics, Merck & Co., Inc, Rahway, NJ, USA
| |
Collapse
|
8
|
Peng J, Yi J, Yang G, Huang Z, Cao D. ISTransbase: an online database for inhibitor and substrate of drug transporters. Database (Oxford) 2024; 2024:baae053. [PMID: 38943608 PMCID: PMC11214160 DOI: 10.1093/database/baae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/17/2024] [Accepted: 06/17/2024] [Indexed: 07/01/2024]
Abstract
Drug transporters, integral membrane proteins found throughout the human body, play critical roles in physiological and biochemical processes through interactions with ligands, such as substrates and inhibitors. The extensive and disparate data on drug transporters complicate understanding their complex relationships with ligands. To address this challenge, it is essential to gather and summarize information on drug transporters, inhibitors and substrates, and simultaneously develop a comprehensive and user-friendly database. Current online resources often provide fragmented information and have limited coverage of drug transporter substrates and inhibitors, highlighting the need for a specialized, comprehensive and openly accessible database. ISTransbase addresses this gap by amassing a substantial amount of data from literature, government documents and open databases. It includes 16 528 inhibitors and 4465 substrates of 163 drug transporters from 18 different species, resulting in a total of 93 841 inhibitor records and 51 053 substrate records. ISTransbase provides detailed insights into drug transporters and their inhibitors/substrates, encompassing transporter and molecule structure, transporter function and distribution, as well as experimental methods and results from transport or inhibition experiments. Furthermore, ISTransbase offers three search strategies that allow users to retrieve drugs and transporters based on multiple selectable constraints, as well as perform checks for drug-drug interactions. Users can also browse and download data. In summary, ISTransbase (https://istransbase.scbdd.com/) serves as a valuable resource for accurately and efficiently accessing information on drug transporter inhibitors and substrates, aiding researchers in exploring drug transporter mechanisms and assisting clinicians in mitigating adverse drug reactions Database URL: https://istransbase.scbdd.com/.
Collapse
Affiliation(s)
- Jinfu Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, No.172 Tongzipo Road, Changsha, Hunan 410031, China
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, Hunan 410031, China
| | - Jiacai Yi
- School of Computer Science, National University of Defense Technology, No.869 Furong Middle Road, Changsha, Hunan 410073, China
| | - Guoping Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, No.172 Tongzipo Road, Changsha, Hunan 410031, China
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, Hunan 410031, China
| | - Zhijun Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Changsha, Hunan 410031, China
- XiangYa School of Medicine, Central South University, No.172 Tongzipo Road, Changsha, Hunan 410031, China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, No.172 Tongzipo Road, Changsha, Hunan 410031, China
| |
Collapse
|
9
|
Piscitelli J, Reddy MB, Wollenberg L, Del Frari L, Gong J, Wood L, Zhang Y, Matschke K, Williams JH. Clinical Evaluation of the Effect of Encorafenib on Bupropion, Rosuvastatin, and Coproporphyrin I and Considerations for Statin Coadministration. Clin Pharmacokinet 2024; 63:483-496. [PMID: 38424308 PMCID: PMC11052825 DOI: 10.1007/s40262-024-01352-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND AND OBJECTIVES Encorafenib is a kinase inhibitor indicated for the treatment of patients with unresectable or metastatic melanoma or metastatic colorectal cancer, respectively, with selected BRAF V600 mutations. A clinical drug-drug interaction (DDI) study was designed to evaluate the effect of encorafenib on rosuvastatin, a sensitive substrate of OATP1B1/3 and breast cancer resistance protein (BCRP), and bupropion, a sensitive CYP2B6 substrate. Coproporphyrin I (CP-I), an endogenous substrate for OATP1B1, was measured in a separate study to deconvolute the mechanism of transporter DDI. METHODS DDI study participants received a single oral dose of rosuvastatin (10 mg) and bupropion (75 mg) on days - 7, 1, and 14 and continuous doses of encorafenib (450 mg QD) and binimetinib (45 mg BID) starting on day 1. The CP-I data were collected from participants in a phase 3 study who received encorafenib (300 mg QD) and cetuximab (400 mg/m2 initial dose, then 250 mg/m2 QW). Pharmacokinetic and pharmacodynamic analysis was performed using noncompartmental and compartmental methods. RESULTS Bupropion exposure was not increased, whereas rosuvastatin Cmax and area under the receiver operating characteristic curve (AUC) increased approximately 2.7 and 1.6-fold, respectively, following repeated doses of encorafenib and binimetinib. Increase in CP-I was minimal, suggesting that the primary effect of encorafenib on rosuvastatin is through BCRP. Categorization of statins on the basis of their metabolic and transporter profile suggests pravastatin would have the least potential for interaction when coadministered with encorafenib. CONCLUSION The results from these clinical studies suggest that encorafenib does not cause clinically relevant CYP2B6 induction or inhibition but is an inhibitor of BCRP and may also inhibit OATP1B1/3 to a lesser extent. Based on these results, it may be necessary to consider switching statins or reducing statin dosage accordingly for coadministration with encorafenib. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT03864042, registered 6 March 2019.
Collapse
|
10
|
Asano S, Kurosaki C, Mori Y, Shigemi R. Quantitative prediction of transporter-mediated drug-drug interactions using the mechanistic static pharmacokinetic (MSPK) model. Drug Metab Pharmacokinet 2024; 54:100531. [PMID: 38064927 DOI: 10.1016/j.dmpk.2023.100531] [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: 05/29/2023] [Revised: 08/21/2023] [Accepted: 10/02/2023] [Indexed: 02/06/2024]
Abstract
Guidance/guidelines on drug-drug interactions (DDIs) have been issued in Japan, the United States, and Europe. These guidance/guidelines provide decision trees for conducting metabolizing enzyme-mediated clinical DDI studies; however, the decision trees for transporter-mediated DDIs lack quantitative prediction methods. In this study, the accuracy of a net-effect mechanistic static pharmacokinetics (MSPK) model containing the fraction transported (ft) of transporters was examined to predict transporter-mediated DDIs. This study collected information on 25 oral drugs with new active reagents that were used in clinical DDI studies as perpetrators (42 cases) from drugs approved in Japan between April 2016 and June 2020. The AUCRs (AUC ratios with and without perpetrators) of victim drugs were predicted using the net-effect MSPK model. As a result, 83 and 95% of the predicted AUCRs were within 1.5- and 2-fold error in the observed AUCRs, respectively. In cases where the victims were statins in which pharmacokinetics several transporters are involved, 70 and 91% of the predicted AUCRs were within 1.5- and 2-fold errors, respectively. Therefore, the net-effect MSPK model was applicable for predicting the AUCRs of victims, which are substrates for multiple transporters.
Collapse
Affiliation(s)
- Satoshi Asano
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; Teijin Pharma Limited, Toxicology & DMPK Development Research Group, 4-3-2, Asahigaoka, Hino, Tokyo, 191-8512, Japan.
| | - Chie Kurosaki
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; FUJIFILM Toyama Chemical Co., Ltd, ADME-Tox Group, Bioanalytical Sciences Research Department, Toyama Research and Development Center, 4-1, Shimo-Okui 2-chome, Toyama-shi, Toyama, Japan
| | - Yuko Mori
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; Pfizer R&D Japan, Clinical Pharmacology and Bioanalytics, Shinjuku Bunka Quint Bldg., 3-22-7, Yoyogi, Shibuya-ku, Tokyo, Japan
| | - Ryota Shigemi
- Japan Pharmaceutical Manufacturers Association, Nihonbashi Life Science Bldg, 2-3-11 Nihonbashi-honcho, Chuo-Ku, Tokyo, Japan; Bayer Yakuhin, Ltd, Preclinical Development, Breeze Tower, 2-4-9, Umeda, Kita-ku, Osaka, Japan
| |
Collapse
|
11
|
Damiani D, Tiribelli M. ATP-Binding Cassette Subfamily G Member 2 in Acute Myeloid Leukemia: A New Molecular Target? Biomedicines 2024; 12:111. [PMID: 38255216 PMCID: PMC10813371 DOI: 10.3390/biomedicines12010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Despite the progress in the knowledge of disease pathogenesis and the identification of many molecular markers as potential targets of new therapies, the cure of acute myeloid leukemia remains challenging. Disease recurrence after an initial response and the development of resistance to old and new therapies account for the poor survival rate and still make allogeneic stem cell transplantation the only curative option. Multidrug resistance (MDR) is a multifactorial phenomenon resulting from host-related characteristics and leukemia factors. Among these, the overexpression of membrane drug transporter proteins belonging to the ABC (ATP-Binding Cassette)-protein superfamily, which diverts drugs from their cellular targets, plays an important role. Moreover, a better understanding of leukemia biology has highlighted that, at least in cancer, ABC protein's role goes beyond simple drug transport and affects many other cell functions. In this paper, we summarized the current knowledge of ABCG2 (formerly Breast Cancer Resistance Protein, BCRP) in acute myeloid leukemia and discuss the potential ways to overcome its efflux function and to revert its ability to confer stemness to leukemia cells, favoring the persistence of leukemia progenitors in the bone marrow niche and justifying relapse also after therapy intensification with allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| |
Collapse
|
12
|
Ronaldson PT, Davis TP. Blood-brain barrier transporters: a translational consideration for CNS delivery of neurotherapeutics. Expert Opin Drug Deliv 2024; 21:71-89. [PMID: 38217410 PMCID: PMC10842757 DOI: 10.1080/17425247.2024.2306138] [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: 10/02/2023] [Accepted: 01/12/2024] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Successful neuropharmacology requires optimization of CNS drug delivery and, by extension, free drug concentrations at brain molecular targets. Detailed assessment of blood-brain barrier (BBB) physiological characteristics is necessary to achieve this goal. The 'next frontier' in CNS drug delivery is targeting BBB uptake transporters, an approach that requires evaluation of brain endothelial cell transport processes so that effective drug accumulation and improved therapeutic efficacy can occur. AREAS COVERED BBB permeability of drugs is governed by tight junction protein complexes (i.e., physical barrier) and transporters/enzymes (i.e., biochemical barrier). For most therapeutics, a component of blood-to-brain transport involves passive transcellular diffusion. Small molecule drugs that do not possess acceptable physicochemical characteristics for passive permeability may utilize putative membrane transporters for CNS uptake. While both uptake and efflux transport mechanisms are expressed at the brain microvascular endothelium, uptake transporters can be targeted for optimization of brain drug delivery and improved treatment of neurological disease states. EXPERT OPINION Uptake transporters represent a unique opportunity to optimize brain drug delivery by leveraging the endogenous biology of the BBB. A rigorous understanding of these transporters is required to improve translation from the bench to clinical trials and stimulate the development of new treatment paradigms for neurological diseases.
Collapse
Affiliation(s)
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona College of Medicine
| |
Collapse
|
13
|
Deng F, Sistonen J, Neuvonen M, Niemi M. Inhibition of efflux transporters by poly ADP-ribose polymerase inhibitors. Basic Clin Pharmacol Toxicol 2023; 133:428-436. [PMID: 37539467 DOI: 10.1111/bcpt.13928] [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: 05/05/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/05/2023]
Abstract
Poly ADP-ribose polymerase (PARP) inhibitors have been approved for the treatment of various cancers. They share a similar mechanism of action but have differences in pharmacokinetic characteristics and potential for drug-drug interactions (DDI). This study evaluated the potential ATP-binding cassette transporter-mediated interactions between PARP inhibitors (niraparib, olaparib and rucaparib) and statins (atorvastatin and rosuvastatin). We studied the inhibitory activity of PARP inhibitors on breast cancer resistance protein (BCRP), multidrug resistance-associated protein 3 (MRP3) and P-glycoprotein (P-gp) using vesicular transport assays and determined the concentrations required for 50% inhibition (IC50 ). Then, we predicted the increase of statin exposure followed by the administration of PARP inhibitors using a mechanistic static model. Rucaparib was the strongest inhibitor of BCRP-mediated rosuvastatin transport (IC50 13.7 μM), followed by niraparib (42.6 μM) and olaparib (216 μM). PARP inhibitors did not affect MRP3. While niraparib appeared to inhibit P-gp, the inhibition showed large variability. The inhibition of intestinal BCRP by rucaparib, niraparib and olaparib was predicted to elevate rosuvastatin exposure by 52%, 37% and 24%, respectively. The interactions between PARP inhibitors and rosuvastatin are probably of minor clinical significance alone, but combined with other predisposing factors, they may increase the risk of rosuvastatin-associated adverse effects.
Collapse
Affiliation(s)
- Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Sistonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
14
|
Chevalier C, Fouqueray P, Bolze S. Imeglimin: A Clinical Pharmacology Review. Clin Pharmacokinet 2023; 62:1393-1411. [PMID: 37713097 DOI: 10.1007/s40262-023-01301-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 09/16/2023]
Abstract
Imeglimin (PXL008, EMD-387008, Twymeeg®) is a first-in-class novel oral hypoglycemic agent, launched in Japan, for the treatment of type 2 diabetes mellitus. Its mechanism of action targets mitochondrial bioenergetics to ameliorate insulin resistance and to enhance β-cell function. This review summarizes the properties underlying the pharmacokinetic profile of imeglimin, a small cationic drug belonging to the tetrahydrotriazine chemical class, with a complex mechanism of absorption involving an active transport through organic cation transporters (OCTs). Imeglimin absorption decreases when dose increases due to the saturation of the active uptake transport. Post absorption, imeglimin is rapidly and primarily distributed to organs and tissues, and has a half-life ranging from 9.03 to 20.2 h. Plasma protein binding of imeglimin is low, which explains the rapid distribution to the organs observed in all species. Imeglimin is excreted unchanged in urine, indicating a low extent of metabolism. Imeglimin is a substrate of multidrug and toxic compound extrusion (MATE) 2-K and a substrate and inhibitor of OCT1, OCT2, and MATE1. Clinical drug-drug interaction studies confirmed the absence of relevant clinical interaction with substrates or inhibitors of these transporters. Overall, the drug-drug interaction potential of imeglimin is low. Its pharmacokinetics profile has also been characterized in special populations, showing no influence of mild and moderate hepatic impairment but an impact of renal function on imeglimin renal clearance. Dosage adjustment is thus required in moderately and severely renally impaired patients. Imeglimin pharmacokinetics was shown to be insensitive to ethnicity and food intake and to have no effect on QTcF interval.
Collapse
|
15
|
Vandecruys P, Baldewijns S, Sillen M, Van Genechten W, Van Dijck P. Oteseconazole: a long-awaited diversification of the antifungal arsenal to manage recurrent vulvovaginal candidiasis (RVVC). Expert Rev Anti Infect Ther 2023; 21:799-812. [PMID: 37449774 DOI: 10.1080/14787210.2023.2233696] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/06/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Recurrent vulvovaginal candidiasis (RVVC) affects women worldwide and has far-reaching implications for a patient's quality of life. For decades, maintenance treatment using the azole antifungal fluconazole was the preferred treatment. Although efficient in controlling the symptoms, the development of azole resistance and high rates of recurrence after therapy cessation have emerged as significant limitations. Nevertheless, persistent efforts have delivered novel treatment options. Oteseconazole (VT-1161), marketed as VIVJOA, is an oral, tetrazole antifungal with unprecedented specificity toward the fungal lanosterol 14α-demethylase. AREAS COVERED We reviewed literature data on oteseconazole with a focus on the management of RVVC. EXPERT OPINION Therapeutic options for RVVC are limited, and novel, innovative approaches are needed to treat this debilitating condition. These therapies need to be well-tolerated and prevent RVVC recurrence. The available clinical data show excellent safety and efficacy, with an unprecedentedly low recurrence rate. However, we believe health-care providers should be mindful to monitor for the development of resistance, as this may result in treatment failure. Further, the availability and cost may, like for most novel drugs, affect the widespread clinical implementation of VIVJOA. Altogether, we are convinced that VIVJOA is a significant advance in RVVC management.
Collapse
Affiliation(s)
- Paul Vandecruys
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Silke Baldewijns
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mart Sillen
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Wouter Van Genechten
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Damiani D, Tiribelli M. ABCG2 in Acute Myeloid Leukemia: Old and New Perspectives. Int J Mol Sci 2023; 24:ijms24087147. [PMID: 37108308 PMCID: PMC10138346 DOI: 10.3390/ijms24087147] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Despite recent advances, prognosis of acute myeloid leukemia (AML) remains unsatisfactory due to poor response to therapy or relapse. Among causes of resistance, over-expression of multidrug resistance (MDR) proteins represents a pivotal mechanism. ABCG2 is an efflux transporter responsible for inducing MDR in leukemic cells; through its ability to extrude many antineoplastic drugs, it leads to AML resistance and/or relapse, even if conflicting data have been reported to date. Moreover, ABCG2 may be co-expressed with other MDR-related proteins and is finely regulated by epigenetic mechanisms. Here, we review the main issues regarding ABCG2 activity and regulation in the AML clinical scenario, focusing on its expression and the role of polymorphisms, as well as on the potential ways to inhibit its function to counteract drug resistance to, eventually, improve outcomes in AML patients.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, P.le Santa Maria della Misericordia, 5, 33100 Udine, Italy
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, P.le Santa Maria della Misericordia, 5, 33100 Udine, Italy
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| |
Collapse
|
17
|
Elsby R, Coghlan H, Edgerton J, Hodgson D, Outteridge S, Atkinson H. Mechanistic in vitro studies indicate that the clinical drug-drug interactions between protease inhibitors and rosuvastatin are driven by inhibition of intestinal BCRP and hepatic OATP1B1 with minimal contribution from OATP1B3, NTCP and OAT3. Pharmacol Res Perspect 2023; 11:e01060. [PMID: 36811234 PMCID: PMC9944867 DOI: 10.1002/prp2.1060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 02/24/2023] Open
Abstract
Previous use of a mechanistic static model to accurately quantify the increased rosuvastatin exposure due to drug-drug interaction (DDI) with coadministered atazanavir underpredicted the magnitude of area under the plasma concentration-time curve ratio (AUCR) based on inhibition of breast cancer resistance protein (BCRP) and organic anion transporting polypeptide (OATP) 1B1. To reconcile the disconnect between predicted and clinical AUCR, atazanavir and other protease inhibitors (darunavir, lopinavir and ritonavir) were evaluated as inhibitors of BCRP, OATP1B1, OATP1B3, sodium taurocholate cotransporting polypeptide (NTCP) and organic anion transporter (OAT) 3. None of the drugs inhibited OAT3, nor did darunavir and ritonavir inhibit OATP1B3 or NTCP. All drugs inhibited BCRP-mediated estrone 3-sulfate transport or OATP1B1-mediated estradiol 17β-D-glucuronide transport with the same rank order of inhibitory potency (lopinavir>ritonavir>atazanavir>>darunavir) and mean IC50 values ranging from 15.5 ± 2.80 μM to 143 ± 14.7 μM or 0.220 ± 0.0655 μM to 9.53 ± 2.50 μM, respectively. Atazanavir and lopinavir also inhibited OATP1B3- or NTCP-mediated transport with a mean IC50 of 1.86 ± 0.500 μM or 65.6 ± 10.7 μM and 5.04 ± 0.0950 μM or 20.3 ± 2.13 μM, respectively. Following integration of a combined hepatic transport component into the previous mechanistic static model using the in vitro inhibitory kinetic parameters determined above for atazanavir, the newly predicted rosuvastatin AUCR reconciled with the clinically observed AUCR confirming additional minor involvement of OATP1B3 and NTCP inhibition in its DDI. The predictions for the other protease inhibitors confirmed inhibition of intestinal BCRP and hepatic OATP1B1 as the principal pathways involved in their clinical DDI with rosuvastatin.
Collapse
Affiliation(s)
- Robert Elsby
- Department of Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec Company)MacclesfieldCheshireUK
| | - Hannah Coghlan
- Department of Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec Company)MacclesfieldCheshireUK
- Present address:
Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety ScienceUniversity of LiverpoolLiverpoolUK
| | - Jacob Edgerton
- Department of Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec Company)MacclesfieldCheshireUK
| | - David Hodgson
- Department of Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec Company)MacclesfieldCheshireUK
| | - Samuel Outteridge
- Department of Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec Company)MacclesfieldCheshireUK
| | - Hayley Atkinson
- Department of Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec Company)MacclesfieldCheshireUK
| |
Collapse
|
18
|
Lee L, Murase K, Ma J, Thoolen M. Clinical Drug-Drug Interaction Between Vatiquinone, a 15-Lipoxygenase Inhibitor, and Rosuvastatin, a Breast Cancer Resistance Protein Substrate. Clin Pharmacol Drug Dev 2023; 12:279-286. [PMID: 36478438 DOI: 10.1002/cpdd.1199] [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: 07/22/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022]
Abstract
Vatiquinone is a small-molecule inhibitor of 15-lipoxygenase in phase 3 development for patients with mitochondrial disease and Friedreich ataxia. The objective of this analysis was to determine the effect of vatiquinone on the pharmacokinetic profile of rosuvastatin, a breast cancer resistance protein substrate. In vitro investigations demonstrated potential inhibition of BCRP by vatiquinone (half maximal inhibitory concentration, 3.8 µM). An open-label, fixed-sequence drug-drug interaction study in healthy volunteers was conducted to determine the clinical relevance of this finding. Subjects received a single dose of 20-mg rosuvastatin followed by a 7-day washout. On days 8 through 14, subjects received 400 mg of vatiquinone 3 times daily. On day 12, subjects concomitantly received a single dose of 20-mg rosuvastatin. The geometric mean ratio for maximum plasma concentration was 77.8%; however, the rosuvastatin disposition phase appeared unaffected. The geometric mean ratios for the area under the plasma concentration-time curve from time 0 to time t and from time 0 to infinity were 103.2% and 99.9%, respectively. Mean rosuvastatin apparent elimination half-life was similar between treatment groups. These results demonstrate that vatiquinone has no clinically relevant effect on the pharmacokinetics of rosuvastatin.
Collapse
Affiliation(s)
- Lucy Lee
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| | | | - Jiyuan Ma
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| | - Martin Thoolen
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| |
Collapse
|
19
|
Sharma S, Mettu VS, Prasad B. Interplay of Breast Cancer Resistance Protein (Bcrp/Abcg2), Sex, and Fed State in Oral Pharmacokinetic Variability of Furosemide in Rats. Pharmaceutics 2023; 15:pharmaceutics15020542. [PMID: 36839862 PMCID: PMC9968170 DOI: 10.3390/pharmaceutics15020542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Poor and variable oral bioavailability of furosemide (FUR) presents critical challenges in pharmacotherapy. We investigated the interplay of breast cancer resistance protein (Bcrp)-mediated transport, sex, and fed state on FUR pharmacokinetics (PK) in rats. A crossover PK study of FUR (5 mg/kg, oral) was performed in Sprague-Dawley rats (3 males and 3 females), alone or with a Bcrp inhibitor, novobiocin (NOV) (20 mg/kg, oral), in both fed and fasted states. Co-administration of NOV significantly increased FUR extent (AUC) and rate (Cmax) of exposure by more than two-fold, which indicates efficient Bcrp inhibition in the intestine. The female rats showed two-fold higher AUC and Cmax, and two-fold lower renal clearance of FUR compared to the male rats. The latter was correlated with higher renal abundance of Bcrp and organic anion transporters (Oats) in the male rats compared to age-matched female rats. These findings suggest that the PK of Bcrp and/or Oat substrates could be sex-dependent in rats. Moreover, allometric scaling of rat PK and toxicological data of Bcrp substrates should consider species and sex differences in Bcrp and Oat abundance in the kidney. Considering that Bcrp is abundant in the intestine of rats and humans, a prospective clinical study is warranted to evaluate the effect of Bcrp inhibition on FUR PK. The potential confounding effect of the Bcrp transporter should be considered when FUR is used as a clinical probe of renal organic anion transporter-mediated drug-drug interactions. Unlike human data, no food-effect was observed on FUR PK in rats.
Collapse
Affiliation(s)
| | | | - Bhagwat Prasad
- Correspondence: ; Tel.: +1-(509)-358-7739; Fax: +1-509-368-6561
| |
Collapse
|
20
|
Deng F, Sjöstedt N, Santo M, Neuvonen M, Niemi M, Kidron H. Novel inhibitors of breast cancer resistance protein (BCRP, ABCG2) among marketed drugs. Eur J Pharm Sci 2023; 181:106362. [PMID: 36529162 DOI: 10.1016/j.ejps.2022.106362] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/11/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Drug-drug interactions (DDIs) are a major concern for the safe use of medications. Breast cancer resistance protein (BCRP) is a clinically relevant ATP-binding cassette (ABC) transporter for drug disposition. Inhibition of BCRP increases the plasma concentrations of BCRP substrate drugs, which potentially could lead to adverse drug reactions. The aim of the present study was to identify BCRP inhibitors amongst a library of 232 commonly used drugs and anticancer drugs approved by the United States Food and Drug Administration (FDA). BCRP inhibition studies were carried out using the vesicular transport assay. We found 75 drugs that reduced the relative transport activity of BCRP to less than 25% of the vehicle control and were categorized as strong inhibitors. The concentration required for 50% inhibition (IC50) was determined for 13 strong inhibitors that were previously poorly characterized for BCRP inhibition. The IC50 ranged from 1.1 to 11 µM, with vemurafenib, dabigatran etexilate and everolimus being the strongest inhibitors. According to the drug interaction guidance documents from the FDA and the European Medicines Agency (EMA), in vivo DDI studies are warranted if the theoretical intestinal luminal concentration of a drug exceeds its IC50 by tenfold. Here, the IC50 values for eight of the drugs were 100-fold lower than their theoretical intestinal luminal concentration. Moreover, a mechanistic static model suggested that vemurafenib, bexarotene, dabigatran etexilate, rifapentine, aprepitant, and ivacaftor could almost fully inhibit intestinal BCRP, increasing the exposure of concomitantly administered rosuvastatin over 90%. Therefore, clinical studies are warranted to investigate whether these drugs cause BCRP-mediated DDIs in humans.
Collapse
Affiliation(s)
- Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland
| | - Noora Sjöstedt
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland
| | - Mariangela Santo
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Heidi Kidron
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland.
| |
Collapse
|
21
|
Naseem A, Pal A, Gowan S, Asad Y, Donovan A, Temesszentandrási-Ambrus C, Kis E, Gaborik Z, Bhalay G, Raynaud F. Intracellular Metabolomics Identifies Efflux Transporter Inhibitors in a Routine Caco-2 Cell Permeability Assay-Biological Implications. Cells 2022; 11:3286. [PMID: 36291153 PMCID: PMC9601193 DOI: 10.3390/cells11203286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 12/05/2023] Open
Abstract
Caco-2 screens are routinely used in laboratories to measure the permeability of compounds and can identify substrates of efflux transporters. In this study, we hypothesized that efflux transporter inhibition of a compound can be predicted by an intracellular metabolic signature in Caco-2 cells in the assay used to test intestinal permeability. Using selective inhibitors and transporter knock-out (KO) cells and a targeted Liquid Chromatography tandem Mass Spectrometry (LC-MS) method, we identified 11 metabolites increased in cells with depleted P-glycoprotein (Pgp) activity. Four metabolites were altered with Breast Cancer Resistance (BCRP) inhibition and nine metabolites were identified in the Multidrug Drug Resistance Protein 2 (MRP2) signature. A scoring system was created that could discriminate among the three transporters and validated with additional inhibitors. Pgp and MRP2 substrates did not score as inhibitors. In contrast, BCRP substrates and inhibitors showed a similar intracellular metabolomic signature. Network analysis of signature metabolites led us to investigate changes of enzymes in one-carbon metabolism (folate and methionine cycles). Our data shows that methylenetetrahydrofolate reductase (MTHFR) protein levels increased with Pgp inhibition and Thymidylate synthase (TS) protein levels were reduced with Pgp and MRP2 inhibition. In addition, the methionine cycle is also affected by both Pgp and MRP2 inhibition. In summary, we demonstrated that the routine Caco-2 assay has the potential to identify efflux transporter inhibitors in parallel with substrates in the assays currently used in many DMPK laboratories and that inhibition of efflux transporters has biological consequences.
Collapse
Affiliation(s)
- Afia Naseem
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Rd., Sutton SM2 5NG, UK
| | - Akos Pal
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Rd., Sutton SM2 5NG, UK
| | - Sharon Gowan
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Rd., Sutton SM2 5NG, UK
| | - Yasmin Asad
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Rd., Sutton SM2 5NG, UK
| | - Adam Donovan
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Rd., Sutton SM2 5NG, UK
| | | | - Emese Kis
- SOLVO Biotechnology, Charles River Company, Irinyi József u. 4-20, 1117 Budapest, Hungary
| | - Zsuzsanna Gaborik
- SOLVO Biotechnology, Charles River Company, Irinyi József u. 4-20, 1117 Budapest, Hungary
| | - Gurdip Bhalay
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Rd., Sutton SM2 5NG, UK
| | - Florence Raynaud
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Rd., Sutton SM2 5NG, UK
| |
Collapse
|
22
|
Elsby R, Atkinson H, Butler P, Riley RJ. Studying the right transporter at the right time: an in vitro strategy for assessing drug-drug interaction risk during drug discovery and development. Expert Opin Drug Metab Toxicol 2022; 18:619-655. [PMID: 36205497 DOI: 10.1080/17425255.2022.2132932] [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: 11/04/2022]
Abstract
INTRODUCTION Transporters are significant in dictating drug pharmacokinetics, thus inhibition of transporter function can alter drug concentrations resulting in drug-drug interactions (DDIs). Because they can impact drug toxicity, transporter DDIs are a regulatory concern for which prediction of clinical effect from in vitro data is critical to understanding risk. AREA COVERED The authors propose in vitro strategies to assist mitigating/removing transporter DDI risk during development by frontloading specific studies, or managing patient risk in the clinic. An overview of clinically relevant drug transporters and observed DDIs are provided, alongside presentation of key considerations/recommendations for in vitro study design evaluating drugs as inhibitors or substrates. Guidance on identifying critical co-medications, clinically relevant disposition pathways and using mechanistic static equations for quantitative prediction of DDI is compiled. EXPERT OPINION The strategies provided will facilitate project teams to study the right transporter at the right time to minimise development risks associated with DDIs. To truly alleviate or manage clinical risk, the industry will benefit from moving away from current qualitative basic static equation approaches to transporter DDI hazard assessment towards adopting the use of mechanistic models to enable quantitative DDI prediction, thereby contextualising risk to ascertain whether a transporter DDI is simply pharmacokinetic or clinically significant requiring intervention.
Collapse
Affiliation(s)
- Robert Elsby
- Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec company), Alderley Park, Macclesfield, Cheshire, United Kingdom
| | - Hayley Atkinson
- Drug Transporter Sciences, Cyprotex Discovery Ltd (an Evotec company), Alderley Park, Macclesfield, Cheshire, United Kingdom
| | - Philip Butler
- ADME Sciences, Cyprotex Discovery Ltd (an Evotec company), Alderley Park, Macclesfield, Cheshire, United Kingdom
| | - Robert J Riley
- Drug Metabolism and Pharmacokinetics, Evotec, Abingdon, Oxfordshire, United Kingdom
| |
Collapse
|
23
|
The next frontier in ADME science: Predicting transporter-based drug disposition, tissue concentrations and drug-drug interactions in humans. Pharmacol Ther 2022; 238:108271. [DOI: 10.1016/j.pharmthera.2022.108271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 12/25/2022]
|
24
|
Matsukane R, Suetsugu K, Hirota T, Ieiri I. Clinical Pharmacokinetics and Pharmacodynamics of Fostamatinib and Its Active Moiety R406. Clin Pharmacokinet 2022; 61:955-972. [PMID: 35781630 PMCID: PMC9250994 DOI: 10.1007/s40262-022-01135-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2022] [Indexed: 11/22/2022]
Abstract
Fostamatinib is the first approved spleen tyrosine kinase inhibitor for chronic immune thrombocytopenia. This review summarizes the clinical development, pharmacokinetics, pharmacodynamics, drug-drug interactions, adverse events, and comprehensive analyses of fostamatinib. While integrating these findings, we discuss the fostering and improvement of fostamatinib for further clinical applications. Fostamatinib is designed as a prodrug and cleavage of its active moiety R406 in the intestine. As R406 is the major product in the blood, this review mainly discusses the pharmacokinetics and pharmacodynamics of R406. It is metabolized by cytochrome 3A4 and UGT1A9 in the liver and is dominantly excreted in feces after anaerobic modification by the gut microbiota. As fostamatinib and R406 strongly inhibit the breast cancer resistance protein, the interaction with those substrates, particularly statins, should be carefully monitored. In patients with immune thrombocytopenia, fostamatinib administration started at 100 mg twice daily, and most patients increased to 150 mg twice daily in the clinical trial. Although responders showed a higher R406 concentration than non-responders, the correlation between R406 exposure and achievement of the platelet count as a pharmacodynamic marker was uncertain in the pharmacokinetic/pharmacodynamic analysis. Additionally, R406 concentration was almost halved in patients with a heavy body weight; hence, the exposure-efficacy study for suitable dosing should be continued with post-marketing data. In contrast, the pharmacokinetic/pharmacodynamic analysis for exposure safety revealed that R406 exposure significantly correlated with the incidence of hypertension. Even though the influence of elevated exposure on other toxicities, including diarrhea and neutropenia, is still unclear, careful management is required with dose escalation to avoid toxicity-related discontinuation.
Collapse
Affiliation(s)
| | | | - Takeshi Hirota
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | - Ichiro Ieiri
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| |
Collapse
|
25
|
Targeting breast cancer resistance protein (BCRP/ABCG2): Functional inhibitors and expression modulators. Eur J Med Chem 2022; 237:114346. [DOI: 10.1016/j.ejmech.2022.114346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/15/2022] [Accepted: 04/01/2022] [Indexed: 12/16/2022]
|
26
|
Groenendaal-van de Meent D, den Adel M, Kerbusch V, van Dijk J, Shibata T, Kato K, Schaddelee M. Effect of Roxadustat on the Pharmacokinetics of Simvastatin, Rosuvastatin, and Atorvastatin in Healthy Subjects: Results From 3 Phase 1, Open-Label, 1-Sequence, Crossover Studies. Clin Pharmacol Drug Dev 2022; 11:486-501. [PMID: 35182045 PMCID: PMC9306950 DOI: 10.1002/cpdd.1076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022]
Abstract
Roxadustat inhibits breast cancer resistance protein and organic anion transporting polypeptide 1B1, which can affect coadministered statin concentrations. Three open‐label, 1‐sequence crossover phase 1 studies in healthy subjects were conducted to assess effects from steady‐state 200‐mg roxadustat on pharmacokinetics and tolerability of 40‐mg simvastatin (CL‐0537 and CL‐0541), 40‐mg atorvastatin (CL‐0538), or 10‐mg rosuvastatin (CL‐0537). Statins were dosed concomitantly with roxadustat in 28 (CL‐0537) and 24 (CL‐0538) healthy subjects, resulting in increases of maximum plasma concentration (Cmax) and area under the plasma concentration–time curve from the time of dosing extrapolated to infinity (AUCinf) 1.87‐ and 1.75‐fold for simvastatin, 2.76‐ and 1.85‐fold for simvastatin acid, 4.47‐ and 2.93‐fold for rosuvastatin, and 1.34‐ and 1.96‐fold for atorvastatin, respectively. Additionally, simvastatin dosed 2 hours before, and 4 and 10 hours after roxadustat in 28 (CL‐0541) healthy subjects, resulted in increases of Cmax and AUCinf 2.32‐ to 3.10‐fold and 1.56‐ to 1.74‐fold for simvastatin and 2.34‐ to 5.98‐fold and 1.89‐ to 3.42‐fold for simvastatin acid, respectively. These increases were not attenuated by time‐separated statin dosing. No clinically relevant differences were observed for terminal elimination half‐life. Concomitant 200‐mg roxadustat and a statin was generally well tolerated during the study period. Roxadustat effects on statin Cmax and AUCinf were statin and administration time dependent. When coadministered with roxadustat, statin‐associated adverse reactions and the need for statin dose reduction should be evaluated.
Collapse
Affiliation(s)
| | | | | | - Jan van Dijk
- Astellas Pharma Europe B.V., Leiden, The Netherlands
| | | | | | | |
Collapse
|
27
|
Merćep I, Radman I, Trkulja V, Božina T, Šimičević L, Budimir E, Ganoci L, Božina N. Loss of function polymorphisms in SLCO1B1 (c.521T>C, rs4149056) and ABCG2 (c.421C>A, rs2231142) genes are associated with adverse events of rosuvastatin: a case-control study. Eur J Clin Pharmacol 2022; 78:227-236. [PMID: 34668025 DOI: 10.1007/s00228-021-03233-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE The study aims to evaluate relationship between polymorphisms associated with a reduced function of two transporter proteins resulting in increased exposure to rosuvastatin - organic anion transporter 1B1 (OATP1B1) (SLCO1B1 c.521T>C) and ATP binding cassette subfamily G member 2 (ABCG2) (ABCG2 c.421C>A) and occurrence of rosuvastatin related myotoxicity/hepatotoxicity. METHODS In a case-control study, cases (rosuvastatin treated patients developing myotoxicity or hepatotoxicity) and controls (concurrent rosuvastatin treated patients free of adverse events) were prospectively recruited over a 2 year period in a single tertiary center specialized in treatment of metabolic disorders. Subjects were evaluated for clinical, comorbidity, and comedication characteristics and for genotype predicted metabolizing phenotypes regarding cytochrome P450 enzymes CYP2C9 and CYP2C19. Standard regression analysis and analysis in matched sets of cases and controls (optimal full matching) were undertaken by fitting frequentist and Bayesian models (covariates/matching variables: age, sex, diabetes, liver/renal disease, hypertension, CYP2C9 and C19 phenotype, use of CYP or transporter inhibitors, non evaluated transporter genotype). RESULTS A total of 88 cases (81 with myotoxicity, 6 with hepatotoxicity, 1 with both) and 129 controls were recruited. Odds of variant SLCO1B1 c.521T>C allele were 2.2-2.5 times higher in cases than in controls (OR = 2.45, 95% CI 1.34-4.48; Bayesian OR = 2.59, 95% CrI 1.42-4.90 in regression analysis; OR = 2.20, 1.10-4.42; Bayesian OR = 2.26, 1.28-4.41 in matched analysis). Odds of variant ABCG2 c.421C>A allele were 2.1-2.3 times higher in cases than in controls (OR = 2.24, 1.04-4.83; Bayesian OR = 2.35, 1.09-4.31 in regression analysis; OR = 2.10, 0.83-5.31; Bayesian OR = 2.17, 1.07-4.35 in matched analysis). CONCLUSION Loss of function polymorphisms in SLCO1B1 c.521T>C and ABCG2 c.421C>A genes are associated with the presence of rosuvastatin related myotoxicity and/or hepatotoxicity.
Collapse
Affiliation(s)
- Iveta Merćep
- Department of Internal Medicine, University Hospital Centre Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivana Radman
- University Department of Ophthalmology, University Hospital Centre Sestre Milosrdnice, Zagreb, Croatia
| | - Vladimir Trkulja
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tamara Božina
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Livija Šimičević
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ema Budimir
- University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lana Ganoci
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nada Božina
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia.
| |
Collapse
|
28
|
Abstract
Circulation of urate levels is determined by the balance between urate production and excretion, homeostasis regulated by the function of urate transporters in key epithelial tissues and cell types. Our understanding of these physiological processes and identification of the genes encoding the urate transporters has advanced significantly, leading to a greater ability to predict risk for urate-associated diseases and identify new therapeutics that directly target urate transport. Here, we review the identified urate transporters and their organization and function in the renal tubule, the intestinal enterocytes, and other important cell types to provide a fuller understanding of the complicated process of urate homeostasis and its role in human diseases. Furthermore, we review the genetic tools that provide an unbiased catalyst for transporter identification as well as discuss the role of transporters in determining the observed significant gender differences in urate-associated disease risk.
Collapse
Affiliation(s)
| | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
29
|
Ning C, Su S, Li J, Kong D, Cai H, Qin Z, Xing H, Chen X, He J. Evaluation of a Clinically Relevant Drug-Drug Interaction Between Rosuvastatin and Clopidogrel and the Risk of Hepatotoxicity. Front Pharmacol 2021; 12:715577. [PMID: 34646133 PMCID: PMC8504577 DOI: 10.3389/fphar.2021.715577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/06/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose: The combination therapy of rosuvastatin (RSV) and the platelet inhibitor clopidogrel (CP) is widely accepted in the management of cardiovascular diseases. The objective of the present study was to identify the mechanism of RSV–CP DDI and evaluate the risk of hepatotoxicity associated with the concomitant use of CP. Methods: We first studied the effect of CP and its major circulating metabolite, carboxylic acid metabolite (CPC), on RSV transport by overexpressing cells and membrane vesicles. Second, we investigated whether a rat model could replicate this DDI and then be used to conduct mechanistic studies and assess the risk of hepatotoxicity. Then, cytotoxicity assay in hepatocytes, biochemical examination, and histopathology were performed to measure the magnitude of liver injury in the presence and absence of DDI. Results: CP inhibited OATP1B1-mediated transport of RSV with an IC50 value of 27.39 μM. CP and CPC inhibited BCRP-mediated RSV transport with IC50 values of <0.001 and 5.96 μM, respectively. The CP cocktail (0.001 μM CP plus 2 μM CPC) significantly inhibited BCRP-mediated transport of RSV by 26.28%. Multiple p.o. doses of CP significantly increased intravenous RSV plasma AUC0-infinity by 76.29% and decreased intravenous RSV CL by 42.62%. Similarly, multiple p.o. doses of CP significantly increased p.o. RSV plasma AUC0-infinity by 87.48% and decreased p.o. RSV CL by 43.27%. CP had no effect on cell viability, while RSV exhibited dose-dependent cytotoxicity after 96 h incubation. Co-incubation of 100 μM CP and RSV for 96 h significantly increased intracellular concentrations and cell-to-medium concentration ratios of RSV and reduced hepatocyte viability. Histological evaluation of liver specimens showed patterns of drug-induced liver injury. Cholestasis was found in rats in the presence of DDI. Conclusion: CP is not a clinically relevant inhibitor for OATP1B1 and OATP1B3. The primary mechanism of RSV–CP DDI can be attributed to the inhibition of intestinal BCRP by CP combined with the inhibition of hepatic BCRP by CPC. The latter is likely to be more clinically relevant and be a contributing factor for increased hepatotoxicity in the presence of DDI.
Collapse
Affiliation(s)
- Chen Ning
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shengdi Su
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiaming Li
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Dexuan Kong
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hui Cai
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhiying Qin
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Han Xing
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiake He
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
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
|
31
|
Tátrai P, Erdő F, Dörnyei G, Krajcsi P. Modulation of Urate Transport by Drugs. Pharmaceutics 2021; 13:pharmaceutics13060899. [PMID: 34204277 PMCID: PMC8235739 DOI: 10.3390/pharmaceutics13060899] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Serum urate (SU) levels in primates are extraordinarily high among mammals. Urate is a Janus-faced molecule that acts physiologically as a protective antioxidant but provokes inflammation and gout when it precipitates at high concentrations. Transporters play crucial roles in urate disposition, and drugs that interact with urate transporters either by intention or by accident may modulate SU levels. We examined whether in vitro transporter interaction studies may clarify and predict such effects. METHODS Transporter interaction profiles of clinically proven urate-lowering (uricosuric) and hyperuricemic drugs were compiled from the literature, and the predictive value of in vitro-derived cut-offs like Cmax/IC50 on the in vivo outcome (clinically relevant decrease or increase of SU) was assessed. RESULTS Interaction with the major reabsorptive urate transporter URAT1 appears to be dominant over interactions with secretory transporters in determining the net effect of a drug on SU levels. In vitro inhibition interpreted using the recommended cut-offs is useful at predicting the clinical outcome. CONCLUSIONS In vitro safety assessments regarding urate transport should be done early in drug development to identify candidates at risk of causing major imbalances. Attention should be paid both to the inhibition of secretory transporters and inhibition or trans-stimulation of reabsorptive transporters, especially URAT1.
Collapse
Affiliation(s)
- Péter Tátrai
- Solvo Biotechnology, Science Park, Building B2, 4-20 Irinyi József utca, H-1117 Budapest, Hungary;
| | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary;
| | - Gabriella Dörnyei
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, H-1088 Budapest, Hungary;
| | - Péter Krajcsi
- Solvo Biotechnology, Science Park, Building B2, 4-20 Irinyi József utca, H-1117 Budapest, Hungary;
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary;
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, H-1088 Budapest, Hungary;
- Correspondence:
| |
Collapse
|
32
|
Mohamed MEF, Coppola S, Feng T, Camp HS, Kim E, Othman AA. Effect of Upadacitinib on the Pharmacokinetics of Rosuvastatin or Atorvastatin in Healthy Subjects. Clin Pharmacol Drug Dev 2021; 10:1335-1344. [PMID: 34109764 PMCID: PMC8596765 DOI: 10.1002/cpdd.957] [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: 01/20/2021] [Accepted: 03/28/2021] [Indexed: 11/08/2022]
Abstract
This phase 1, 2‐part, 2‐period, open‐label, drug‐drug interaction study evaluated the potential for pharmacokinetic interactions between upadacitinib and rosuvastatin, an organic anion transporting polypeptide (OATP) 1B1 and breast cancer resistance protein substrate, or atorvastatin, a cytochrome P450 3A, OATP1B1, and OATP1B3 substrate, in 36 healthy volunteers. During period 1, a single dose of rosuvastatin (5 mg; part 1) or atorvastatin (10 mg; part 2) was administered on day 1, followed by a washout period of 5 days. During period 2, once‐daily doses of upadacitinib extended‐release (30 mg) were administered on days 1 to 10, and a single dose of rosuvastatin (5 mg; part 1) or atorvastatin (10 mg; part 2) was administered 1 hour after the upadacitinib dose on day 7. Serial blood samples were collected for assays of drug concentrations. In Part 1, rosuvastatin maximum observed plasma concentration (Cmax) and area under the plasma concentration–time curve from time 0 to infinity (AUCinf) were 23% and 33% lower, respectively, when administered with upadacitinib relative to when administered alone. In part 2, atorvastatin Cmax and AUCinf was 11% and 23% lower, respectively, when administered with upadacitinib relative to when administered alone. The Cmax and AUCinf of the active metabolite ortho‐hydroxyatorvastatin remained unchanged. Administration of a single 5‐mg dose of rosuvastatin or a single 10‐mg dose of atorvastatin had no relevant effect on upadacitinib Cmax or area under the plasma concentration–time curve. These results demonstrated that upadacitinib has no clinically relevant effect on the pharmacokinetics of rosuvastatin and atorvastatin or on substrates transported by OATP1B or breast cancer resistance protein.
Collapse
Affiliation(s)
| | | | - Tian Feng
- AbbVie Inc., North Chicago, Illinois, USA
| | | | - Elaine Kim
- AbbVie Inc., North Chicago, Illinois, USA
| | | |
Collapse
|
33
|
Mamidi RNVS, Devineni D, Sun D, Yavin Y, Rosenthal N. Rosuvastatin Myotoxicity After Starting Canagliflozin Treatment. Ann Intern Med 2021; 174:431-432. [PMID: 33721528 DOI: 10.7326/l20-1455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | - Don Sun
- Janssen Research & Development, Raritan, New Jersey
| | - Yshai Yavin
- Janssen Research & Development, Raritan, New Jersey
| | | |
Collapse
|
34
|
Auxtero MD, Chalante S, Abade MR, Jorge R, Fernandes AI. Potential Herb-Drug Interactions in the Management of Age-Related Cognitive Dysfunction. Pharmaceutics 2021; 13:124. [PMID: 33478035 PMCID: PMC7835864 DOI: 10.3390/pharmaceutics13010124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Late-life mild cognitive impairment and dementia represent a significant burden on healthcare systems and a unique challenge to medicine due to the currently limited treatment options. Plant phytochemicals have been considered in alternative, or complementary, prevention and treatment strategies. Herbals are consumed as such, or as food supplements, whose consumption has recently increased. However, these products are not exempt from adverse effects and pharmacological interactions, presenting a special risk in aged, polymedicated individuals. Understanding pharmacokinetic and pharmacodynamic interactions is warranted to avoid undesirable adverse drug reactions, which may result in unwanted side-effects or therapeutic failure. The present study reviews the potential interactions between selected bioactive compounds (170) used by seniors for cognitive enhancement and representative drugs of 10 pharmacotherapeutic classes commonly prescribed to the middle-aged adults, often multimorbid and polymedicated, to anticipate and prevent risks arising from their co-administration. A literature review was conducted to identify mutual targets affected (inhibition/induction/substrate), the frequency of which was taken as a measure of potential interaction. Although a limited number of drugs were studied, from this work, interaction with other drugs affecting the same targets may be anticipated and prevented, constituting a valuable tool for healthcare professionals in clinical practice.
Collapse
Affiliation(s)
- Maria D. Auxtero
- CiiEM, Interdisciplinary Research Centre Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal; (M.D.A.); (S.C.); (M.R.A.); (R.J.)
| | - Susana Chalante
- CiiEM, Interdisciplinary Research Centre Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal; (M.D.A.); (S.C.); (M.R.A.); (R.J.)
| | - Mário R. Abade
- CiiEM, Interdisciplinary Research Centre Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal; (M.D.A.); (S.C.); (M.R.A.); (R.J.)
| | - Rui Jorge
- CiiEM, Interdisciplinary Research Centre Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal; (M.D.A.); (S.C.); (M.R.A.); (R.J.)
- Polytechnic Institute of Santarém, School of Agriculture, Quinta do Galinheiro, 2001-904 Santarém, Portugal
- CIEQV, Life Quality Research Centre, IPSantarém/IPLeiria, Avenida Dr. Mário Soares, 110, 2040-413 Rio Maior, Portugal
| | - Ana I. Fernandes
- CiiEM, Interdisciplinary Research Centre Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal; (M.D.A.); (S.C.); (M.R.A.); (R.J.)
| |
Collapse
|
35
|
Parmar HS, Nayak A, Gavel PK, Jha HC, Bhagwat S, Sharma R. Cross Talk between COVID-19 and Breast Cancer. Curr Cancer Drug Targets 2021; 21:575-600. [PMID: 33593260 DOI: 10.2174/1568009621666210216102236] [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: 09/11/2020] [Revised: 12/17/2020] [Accepted: 12/26/2020] [Indexed: 01/08/2023]
Abstract
Cancer patients are more susceptible to COVID-19; however, the prevalence of COVID-19 in different types of cancer is still inconsistent and inconclusive. Here, we delineate the intricate relationship between breast cancer and COVID-19. Breast cancer and COVID-19 share the involvement of common comorbidities, hormonal signalling pathways, gender differences, rennin- angiotensin system (RAS), angiotensin-converting enzyme-2 (ACE-2), transmembrane protease serine 2 (TMPRSS2) and dipeptidyl peptidase-IV (DPP-IV). We also shed light on the possible effects of therapeutic modalities of COVID-19 on breast cancer outcomes. Briefly, we conclude that breast cancer patients are more susceptible to COVID-19 in comparison with their normal counterparts. Women are more resistant to the occurrence and severity of COVID-19. Increased expressions of ACE2 and TMPRSS2 are correlated with occurrence and severity of COVID-19, but higher expression of ACE2 and lower expression of TMPRSS2 are prognostic markers for overall disease free survival in breast cancer. The ACE2 inhibitors and ibuprofen therapies for COVID-19 treatment may aggravate the clinical condition of breast cancer patients through chemo-resistance and metastasis. Most of the available therapeutic modalities for COVID-19 were also found to exert positive effects on breast cancer outcomes. Besides drugs in clinical trend, TMPRSS2 inhibitors, estrogen supplementation, androgen deprivation and DPP-IV inhibitors may also be used to treat breast cancer patients infected with SARS-CoV-2. However, drug-drug interactions suggest that some of the drugs used for the treatment of COVID-19 may modulate the drug metabolism of anticancer therapies which may lead to adverse drug reaction events.
Collapse
Affiliation(s)
| | - Aakruti Nayak
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Pramod Kumar Gavel
- Department of Chemical Sciences, IIT, Indore, Simrol, Indore, M.P., India
| | - Hem Chandra Jha
- Department of Bioscience and Bioengineering, IIT, Indore, Simrol, Indore, M.P., India
| | - Shivani Bhagwat
- Suraksha Diagnostics Pvt. Ltd., Newtown, Rajarhat, Kolkata-West Bengal, India
| | - Rajesh Sharma
- School of Pharmacy, Devi Ahilya University, Indore-452001., M.P., India
| |
Collapse
|
36
|
Al‐Majdoub ZM, Achour B, Couto N, Howard M, Elmorsi Y, Scotcher D, Alrubia S, El‐Khateeb E, Vasilogianni A, Alohali N, Neuhoff S, Schmitt L, Rostami‐Hodjegan A, Barber J. Mass spectrometry-based abundance atlas of ABC transporters in human liver, gut, kidney, brain and skin. FEBS Lett 2020; 594:4134-4150. [PMID: 33128234 PMCID: PMC7756589 DOI: 10.1002/1873-3468.13982] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022]
Abstract
ABC transporters (ATP-binding cassette transporter) traffic drugs and their metabolites across membranes, making ABC transporter expression levels a key factor regulating local drug concentrations in different tissues and individuals. Yet, quantification of ABC transporters remains challenging because they are large and low-abundance transmembrane proteins. Here, we analysed 200 samples of crude and membrane-enriched fractions from human liver, kidney, intestine, brain microvessels and skin, by label-free quantitative mass spectrometry. We identified 32 (out of 48) ABC transporters: ABCD3 was the most abundant in liver, whereas ABCA8, ABCB2/TAP1 and ABCE1 were detected in all tissues. Interestingly, this atlas unveiled that ABCB2/TAP1 may have TAP2-independent functions in the brain and that biliary atresia (BA) and control livers have quite different ABC transporter profiles. We propose that meaningful biological information can be derived from a direct comparison of these data sets.
Collapse
Affiliation(s)
- Zubida M. Al‐Majdoub
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Narciso Couto
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Martyn Howard
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Yasmine Elmorsi
- Clinical Pharmacy DepartmentFaculty of PharmacyTanta UniversityEgypt
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Sarah Alrubia
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Pharmaceutical Chemistry DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Eman El‐Khateeb
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Clinical Pharmacy DepartmentFaculty of PharmacyTanta UniversityEgypt
| | | | - Noura Alohali
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Pharmaceutical Practice DepartmentCollege of PharmacyPrincess Noura Bint Abdul Rahman UniversityRiyadhSaudi Arabia
| | | | - Lutz Schmitt
- Institute of BiochemistryHeinrich Heine University DüsseldorfGermany
| | - Amin Rostami‐Hodjegan
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Simcyp DivisionCertara UK LtdSheffieldUK
| | - Jill Barber
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| |
Collapse
|
37
|
Sane R, Cheung KWK, Kovács P, Farasyn T, Li R, Bui A, Musib L, Kis E, Plise E, Gáborik Z. Calibrating the In Vitro-In Vivo Correlation for OATP-Mediated Drug-Drug Interactions with Rosuvastatin Using Static and PBPK Models. Drug Metab Dispos 2020; 48:1264-1270. [PMID: 33037044 DOI: 10.1124/dmd.120.000149] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/28/2020] [Indexed: 02/13/2025] Open
Abstract
Organic anion-transporting polypeptide (OATP) 1B1/3-mediated drug-drug interaction (DDI) potential is evaluated in vivo with rosuvastatin (RST) as a probe substrate in clinical studies. We calibrated our assay with RST and estradiol 17-β-D-glucuronide (E217βG)/cholecystokinin-8 (CCK8) as in vitro probes for qualitative and quantitative prediction of OATP1B-mediated DDI potential for RST. In vitro OATP1B1/1B3 inhibition using E217βG and CCK8 yielded higher area under the curve (AUC) ratio (AUCR) values numerically with the static model, but all probes performed similarly from a qualitative cutoff-based prediction, as described in regulatory guidances. However, the magnitudes of DDI were not captured satisfactorily. Considering that clearance of RST is also mediated by gut breast cancer resistance protein (BCRP), inhibition of BCRP was also incorporated in the DDI prediction if the gut inhibitor concentrations were 10 × IC50 for BCRP inhibition. This combined static model closely predicted the magnitude of RST DDI with root-mean-square error values of 0.767-0.812 and 1.24-1.31 with and without BCRP inhibition, respectively, for in vitro-in vivo correlation of DDI. Physiologically based pharmacokinetic (PBPK) modeling was also used to simulate DDI between RST and rifampicin, asunaprevir, and velpatasvir. Predicted AUCR for rifampicin and asunaprevir was within 1.5-fold of that observed, whereas that for velpatasvir showed a 2-fold underprediction. Overall, the combined static model incorporating both OATP1B and BCRP inhibition provides a quick and simple mathematical approach to quantitatively predict the magnitude of transporter-mediated DDI for RST for routine application. PBPK complements the static model and provides a framework for studying molecules when a dynamic model is needed. SIGNIFICANCE STATEMENT: Using 22 drugs, we show that a static model for organic anion-transporting polypeptide (OATP) 1B1/1B3 inhibition can qualitatively predict potential for drug-drug interaction (DDI) using a cutoff-based approach, as in regulatory guidances. However, consideration of both OATP1B1/3 and gut breast cancer resistance protein inhibition provided a better prediction of the magnitude of the transporter-mediated DDI of these inhibitors with rosuvastatin. Based on these results, we have proposed an empirical mechanistic-static approach for a more reliable prediction of transporter-mediated DDI liability with rosuvastatin that drug development teams can leverage.
Collapse
Affiliation(s)
- Rucha Sane
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Kit Wun Kathy Cheung
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Péter Kovács
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Taleah Farasyn
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Ruina Li
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Annamaria Bui
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Luna Musib
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Emese Kis
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Emile Plise
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| | - Zsuzsanna Gáborik
- Departments of Clinical Pharmacology (R.S., K.W.K.C., T.F., L.M.) and Drug Metabolism and Pharmacokinetics (T.F., R.L., E.P.), Genentech, Inc., South San Francisco, California; and SOLVO Biotechnology, Budapest, Hungary (P.K., A.B., E.K., Z.G.)
| |
Collapse
|
38
|
Improved In Vitro-In Vivo Correlation by Using the Unbound-Fraction-Adjusted IC 50 for Breast Cancer Resistance Protein Inhibition. Pharm Res 2020; 37:230. [PMID: 33123823 DOI: 10.1007/s11095-020-02954-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/09/2020] [Indexed: 01/16/2023]
Abstract
PURPOSE One function of the blood-brain barrier (BBB) is the efflux of xenobiotics by breast cancer resistance protein (BCRP), and inhibition of BCRP can cause unexpected central nervous system toxicity. Despite the importance of BCRP inhibition and the associated risk of BBB penetration in vivo, there has been little investigation of it to date. In this study, inhibition of BCRP-mediated transport was assessed by in vitro assay in the presence of bovine serum albumin (BSA) to change the unbound inhibitor concentrations, and the in vitro-in vivo correlation (IVIVC) at the BBB was evaluated. METHODS AND RESULTS The IC50 values of BCRP inhibitors were determined in vitro with and without BSA and the inhibitors were categorized into two groups. One group of compounds had little risk of inhibiting BCRP because of their low unbound concentrations. In contrast, the other group has the potential to facilitate BBB penetration by inhibiting BCRP. In the IVIVC approach, brain concentrations and the brain-to-plasma ratio were better correlated with the ratio of the unbound plasma concentration at steady-state to the unbound-fraction-adjusted IC50. CONCLUSION We have found a way to obtain a better in vitro-in vivo correlation for BCRP-mediated transport.
Collapse
|
39
|
Garrison DA, Talebi Z, Eisenmann ED, Sparreboom A, Baker SD. Role of OATP1B1 and OATP1B3 in Drug-Drug Interactions Mediated by Tyrosine Kinase Inhibitors. Pharmaceutics 2020; 12:E856. [PMID: 32916864 PMCID: PMC7559291 DOI: 10.3390/pharmaceutics12090856] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022] Open
Abstract
Failure to recognize important features of a drug's pharmacokinetic characteristics is a key cause of inappropriate dose and schedule selection, and can lead to reduced efficacy and increased rate of adverse drug reactions requiring medical intervention. As oral chemotherapeutic agents, tyrosine kinase inhibitors (TKIs) are particularly prone to cause drug-drug interactions as many drugs in this class are known or suspected to potently inhibit the hepatic uptake transporters OATP1B1 and OATP1B3. In this article, we provide a comprehensive overview of the published literature and publicly-available regulatory documents in this rapidly emerging field. Our findings indicate that, while many TKIs can potentially inhibit the function of OATP1B1 and/or OATP1B3 and cause clinically-relevant drug-drug interactions, there are many inconsistencies between regulatory documents and the published literature. Potential explanations for these discrepant observations are provided in order to assist prescribing clinicians in designing safe and effective polypharmacy regimens, and to provide researchers with insights into refining experimental strategies to further predict and define the translational significance of TKI-mediated drug-drug interactions.
Collapse
Affiliation(s)
| | | | | | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (D.A.G.); (Z.T.); (E.D.E.)
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (D.A.G.); (Z.T.); (E.D.E.)
| |
Collapse
|
40
|
Lee CH, Franchi F, Angiolillo DJ. Clopidogrel drug interactions: a review of the evidence and clinical implications. Expert Opin Drug Metab Toxicol 2020; 16:1079-1096. [PMID: 32835535 DOI: 10.1080/17425255.2020.1814254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Patients with cardiovascular disease are commonly affected by a number of comorbidities leading to a high prevalence of polypharmacy. Polypharmacy increases the probability of drug-drug interactions (DDIs). Amongst these, DDIs involving clopidogrel, the most commonly utilized platelet P2Y12 inhibitor, is a topic of potential clinical concern. AREAS COVERED This article reviews DDIs between clopidogrel and drugs which are widely used in clinical practice. In particular, drugs shown to interfere with the pharmacodynamic and pharmacokinetic effects of clopidogrel and the clinical implications of these findings are reviewed. These drugs include inhibitors of gastric acid secretion, statins, calcium channel blockers, antidiabetic agents, and antimicrobial agents. For the references, we searched PubMed, EMBASE, or the Cochrane Library. EXPERT OPINION Clopidogrel-drug interactions are common. Most of these DDIs are limited to laboratory findings showing an impact on clopidogrel-induced antiplatelet effects. While variability in clopidogrel-induced antiplatelet effects is known to affect clinical outcomes, with high platelet reactivity being associated with thrombotic complications among patients undergoing coronary stenting, most studies assessing the clinical implications of clopidogrel-drug interactions have not shown to significantly affect outcomes. However, awareness of these DDIs remains important for optimizing the selection of concomitant therapies.
Collapse
Affiliation(s)
- Chang Hoon Lee
- Division of Cardiology, University of Florida College of Medicine-Jacksonville , Jacksonville, FL, USA.,Division of Cardiology, Department of Internal Medicine, Veterans Health Service Medical Center , Seoul, Korea
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine-Jacksonville , Jacksonville, FL, USA
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine-Jacksonville , Jacksonville, FL, USA
| |
Collapse
|
41
|
Lack of Drug-Drug Interaction Between Cimetidine, a Renal Transporter Inhibitor, and Imeglimin, a Novel Oral Antidiabetic Drug, in Healthy Volunteers. Eur J Drug Metab Pharmacokinet 2020; 45:725-733. [PMID: 32860624 DOI: 10.1007/s13318-020-00642-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND AND OBJECTIVE: Imeglimin is a novel oral antidiabetic drug to treat type 2 diabetes, targeting the mitochondrial bioenergetics. In vitro, imeglimin was shown to be a substrate of human multidrug and toxic extrusion transporters MATE1 and MATE2-K and organic cation transporters OCT1 and OCT2. The objective of the study was to assess the potential drug-drug interaction between imeglimin and cimetidine, a reference inhibitor of these transporters. METHODS A phase 1 study was carried out in 16 subjects who received a single dose of 1500 mg imeglimin alone on day 1 followed by a 6-day treatment (day 5 to day 10) with cimetidine 400 mg twice daily. On day 8, a single dose of imeglimin was co-administered with cimetidine. Blood and urine samples were collected up to 72 h after each imeglimin administration. Pharmacokinetic parameters were determined using non-compartmental methods. RESULTS Imeglimin maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) were 1.3-fold [90% CI (1.12-1.62) and (1.10-1.46) for Cmax and AUC0-last, respectively] higher when imeglimin was co-administered with cimetidine but this increase was not considered clinically relevant. This increase could be mainly explained by a reduction in renal elimination, mediated through the cimetidine inhibition of renal MATE1 transporter. Imeglimin taken alone or with cimetidine was safe and well tolerated in all subjects. CONCLUSIONS No clinically significant drug-drug interaction exists between imeglimin and cimetidine, a reference inhibitor of MATE1, MATE2-K, OCT1 and OCT2 transporters. CLINICAL TRIAL REGISTRATION EudraCT 2018-001103-36.
Collapse
|
42
|
Hirota T, Fujita Y, Ieiri I. An updated review of pharmacokinetic drug interactions and pharmacogenetics of statins. Expert Opin Drug Metab Toxicol 2020; 16:809-822. [PMID: 32729746 DOI: 10.1080/17425255.2020.1801634] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Hydroxymethylglutaryl-coenzyme A reductase inhibitors (statins) lower cholesterol synthesis in patients with hypercholesterolemia. Increased statin exposure is an important risk factor for skeletal muscle toxicity. Potent inhibitors of cytochrome P450 (CYP) 3A4 significantly increase plasma concentrations of the active forms of simvastatin, lovastatin, and atorvastatin. Fluvastatin is metabolized by CYP2C9, whereas pravastatin, rosuvastatin, and pitavastatin are unaffected by inhibition by either CYP. Statins also have different affinities for membrane transporters involved in processes such as intestinal absorption, hepatic absorption, biliary excretion, and renal excretion. AREAS COVERED In this review, the pharmacokinetic aspects of drug-drug interactions with statins and genetic polymorphisms of CYPs and drug transporters involved in the pharmacokinetics of statins are discussed. EXPERT OPINION Understanding the mechanisms underlying statin interactions can help minimize drug interactions and reduce the adverse side effects caused by statins. Since recent studies have shown the involvement of drug transporters such as OATP and BCRP as well as CYPs in statin pharmacokinetics, further clinical studies focusing on the drug transporters are necessary. The establishment of biomarkers based on novel mechanisms, such as the leakage of microRNAs into the peripheral blood associated with the muscle toxicity, is important for the early detection of statin side effects.
Collapse
Affiliation(s)
- Takeshi Hirota
- Department of Clinical Pharmacokinetics, Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Kyushu University , Fukuoka, Japan
| | - Yuito Fujita
- Department of Clinical Pharmacokinetics, Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Kyushu University , Fukuoka, Japan
| | - Ichiro Ieiri
- Department of Clinical Pharmacokinetics, Division of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Kyushu University , Fukuoka, Japan
| |
Collapse
|
43
|
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
|
44
|
Interpretation of Drug Interaction Using Systemic and Local Tissue Exposure Changes. Pharmaceutics 2020; 12:pharmaceutics12050417. [PMID: 32370191 PMCID: PMC7284846 DOI: 10.3390/pharmaceutics12050417] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022] Open
Abstract
Systemic exposure of a drug is generally associated with its pharmacodynamic (PD) effect (e.g., efficacy and toxicity). In this regard, the change in area under the plasma concentration-time curve (AUC) of a drug, representing its systemic exposure, has been mainly considered in evaluation of drug-drug interactions (DDIs). Besides the systemic exposure, the drug concentration in the tissues has emerged as a factor to alter the PD effects. In this review, the status of systemic exposure, and/or tissue exposure changes in DDIs, were discussed based on the recent reports dealing with transporters and/or metabolic enzymes mediating DDIs. Particularly, the tissue concentration in the intestine, liver and kidney were referred to as important factors of PK-based DDIs.
Collapse
|
45
|
Cui Y, Claus S, Schnell D, Runge F, MacLean C. In-Depth Characterization of EpiIntestinal Microtissue as a Model for Intestinal Drug Absorption and Metabolism in Human. Pharmaceutics 2020; 12:pharmaceutics12050405. [PMID: 32354111 PMCID: PMC7284918 DOI: 10.3390/pharmaceutics12050405] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/31/2022] Open
Abstract
The Caco-2 model is a well-accepted in vitro model for the estimation of fraction absorbed in human intestine. Due to the lack of cytochrome P450 3A4 (CYP3A4) activities, Caco-2 model is not suitable for the investigation of intestinal first-pass metabolism. The purpose of this study is to evaluate a new human intestine model, EpiIntestinal microtissues, as a tool for the prediction of oral absorption and metabolism of drugs in human intestine. The activities of relevant drug transporters and drug metabolizing enzymes, including MDR1 P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), CYP3A4, CYP2J2, UDP-glucuronosyltransferases (UGT), carboxylesterases (CES), etc., were detected in functional assays with selective substrates and inhibitors. Compared to Caco-2, EpiIntestinal microtissues proved to be a more holistic model for the investigation of drug absorption and metabolism in human gastrointestinal tract.
Collapse
Affiliation(s)
- Yunhai Cui
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany; (D.S.); (F.R.)
- Correspondence: ; Tel.: +49-7351-54-92193
| | - Stephanie Claus
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany; (S.C.); (C.M.)
| | - David Schnell
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany; (D.S.); (F.R.)
| | - Frank Runge
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany; (D.S.); (F.R.)
| | - Caroline MacLean
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany; (S.C.); (C.M.)
| |
Collapse
|
46
|
Carter SJ, Chouhan B, Sharma P, Chappell MJ. Prediction of Clinical Transporter-Mediated Drug-Drug Interactions via Comeasurement of Pitavastatin and Eltrombopag in Human Hepatocyte Models. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:211-221. [PMID: 32142598 PMCID: PMC7179958 DOI: 10.1002/psp4.12505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/27/2020] [Indexed: 11/21/2022]
Abstract
A structurally identifiable micro‐rate constant mechanistic model was used to describe the interaction between pitavastatin and eltrombopag, with improved goodness‐of‐fit values through comeasurement of pitavastatin and eltrombopag. Transporter association and dissociation rate constants and passive rates out of the cell were similar between pitavastatin and eltrombopag. Translocation into the cell through transporter‐mediated uptake was six times greater for pitavastatin, leading to pronounced inhibition of pitavastatin uptake by eltrombopag. The passive rate into the cell was 91 times smaller for pitavastatin compared with eltrombopag. A semimechanistic physiologically‐based pharmacokinetic (PBPK) model was developed to evaluate the potential for clinical drug–drug interactions (DDIs). The PBPK model predicted a twofold increase in the pitavastatin peak blood concentration and area under the concentration‐time curve in the presence of eltrombopag in simulated healthy volunteers. The use of structural identifiability supporting experimental design combined with robust micro‐rate constant parameter estimates and a semimechanistic PBPK model gave more informed predictions of transporter‐mediated DDIs.
Collapse
Affiliation(s)
- Simon J Carter
- Biomedical and Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| | - Bhavik Chouhan
- Functional & Mechanistic Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca R&D, Gothenburg, Sweden
| | - Pradeep Sharma
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca R&D, Cambridge, UK
| | - Michael J Chappell
- Biomedical and Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| |
Collapse
|
47
|
Fogli S, Porta C, Del Re M, Crucitta S, Gianfilippo G, Danesi R, Rini BI, Schmidinger M. Optimizing treatment of renal cell carcinoma with VEGFR-TKIs: a comparison of clinical pharmacology and drug-drug interactions of anti-angiogenic drugs. Cancer Treat Rev 2020; 84:101966. [PMID: 32044644 DOI: 10.1016/j.ctrv.2020.101966] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Anti-angiogenic treatment is an important option that has changed the therapeutic landscape in various tumors, particularly in patients affected by renal cell carcinoma (RCC). Agents that block signaling pathways governing tumor angiogenesis have raised high expectations among clinicians. Vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) comprise a heterogeneous class of drugs with distinct pharmacological profiles, including potency, selectivity, pharmacokinetics and drug-drug interactions. Among them, tivozanib is one of the last TKIs introduced in the clinical practice; this drug selectively targets VEGFRs, it is characterized by a favorable pharmacokinetics and safety profile and has been approved as first-line treatment for patients with metastatic RCC (mRCC). In this article, we describe the clinical pharmacology of selected VEGFR-TKIs used for the treatment of mRCC, highlighting the relevant differences; moreover we aim to define the main pharmacologic characteristics of these drug.
Collapse
Affiliation(s)
- Stefano Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Camillo Porta
- Department of Internal Medicine, University of Pavia and Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giulia Gianfilippo
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Brian I Rini
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Manuela Schmidinger
- Clinical Division of Oncology, Department of Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
48
|
Taskar KS, Pilla Reddy V, Burt H, Posada MM, Varma M, Zheng M, Ullah M, Emami Riedmaier A, Umehara KI, Snoeys J, Nakakariya M, Chu X, Beneton M, Chen Y, Huth F, Narayanan R, Mukherjee D, Dixit V, Sugiyama Y, Neuhoff S. Physiologically-Based Pharmacokinetic Models for Evaluating Membrane Transporter Mediated Drug-Drug Interactions: Current Capabilities, Case Studies, Future Opportunities, and Recommendations. Clin Pharmacol Ther 2019; 107:1082-1115. [PMID: 31628859 PMCID: PMC7232864 DOI: 10.1002/cpt.1693] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling has been extensively used to quantitatively translate in vitro data and evaluate temporal effects from drug-drug interactions (DDIs), arising due to reversible enzyme and transporter inhibition, irreversible time-dependent inhibition, enzyme induction, and/or suppression. PBPK modeling has now gained reasonable acceptance with the regulatory authorities for the cytochrome-P450-mediated DDIs and is routinely used. However, the application of PBPK for transporter-mediated DDIs (tDDI) in drug development is relatively uncommon. Because the predictive performance of PBPK models for tDDI is not well established, here, we represent and discuss examples of PBPK analyses included in regulatory submission (the US Food and Drug Administration (FDA), the European Medicines Agency (EMA), and the Pharmaceuticals and Medical Devices Agency (PMDA)) across various tDDIs. The goal of this collaborative effort (involving scientists representing 17 pharmaceutical companies in the Consortium and from academia) is to reflect on the use of current databases and models to address tDDIs. This challenges the common perceptions on applications of PBPK for tDDIs and further delves into the requirements to improve such PBPK predictions. This review provides a reflection on the current trends in PBPK modeling for tDDIs and provides a framework to promote continuous use, verification, and improvement in industrialization of the transporter PBPK modeling.
Collapse
Affiliation(s)
- Kunal S Taskar
- GlaxoSmithKline, DMPK, In Vitro In Vivo Translation, GSK R&D, Ware, UK
| | - Venkatesh Pilla Reddy
- AstraZeneca, Modelling and Simulation, Early Oncology DMPK, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Howard Burt
- Simcyp-Division, Certara UK Ltd., Sheffield, UK
| | | | | | - Ming Zheng
- Bristol-Myers Squibb Company, Princeton, New Jersey, USA
| | | | | | | | - Jan Snoeys
- Janssen Research and Development, Beerse, Belgium
| | | | - Xiaoyan Chu
- Merck Sharp & Dohme Corp., Kenilworth, New Jersey, USA
| | | | - Yuan Chen
- Genentech, San Francisco, California, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Murata H, Ito S, Kusuhara H, Nomura Y, Taniguchi T. Proposal of a Parameter for OATP1B1 Inhibition Screening at the Early Drug Discovery Stage. J Pharm Sci 2019; 108:3898-3902. [DOI: 10.1016/j.xphs.2019.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/29/2019] [Accepted: 08/15/2019] [Indexed: 11/15/2022]
|
50
|
He J, Yu Y, Yin C, Liu H, Zou H, Ma J, Yang W, Liu Y, Zhong L, Chen X. Clinically significant drug‐drug interaction between tacrolimus and fluconazole in stable renal transplant recipient and literature review. J Clin Pharm Ther 2019; 45:264-269. [PMID: 31756280 DOI: 10.1111/jcpt.13075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/05/2019] [Accepted: 10/22/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Jiake He
- Department of Pharmacy The Second Affiliated Hospital of Nanchang University Nanchang China
- Department of Organ Transplantation The Second Affiliated Hospital of Nanchang University Nanchang China
- Clinical Pharmacokinetics Laboratory School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing China
| | - Yang Yu
- Department of Pharmacy The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Chenglong Yin
- Department of Organ Transplantation The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Hailang Liu
- Department of Organ Transplantation The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Hua Zou
- Department of Organ Transplantation The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Jingsheng Ma
- Department of Organ Transplantation The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Wentao Yang
- Department of Organ Transplantation The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Ye Liu
- Department of Pharmacy The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Lin Zhong
- Department of Organ Transplantation The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing China
| |
Collapse
|