1
|
Hagenbuch B, Stieger B, Locher KP. Organic anion transporting polypeptides: Pharmacology, toxicology, structure, and transport mechanisms. Pharmacol Rev 2025; 77:100023. [PMID: 40148036 DOI: 10.1016/j.pharmr.2024.100023] [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: 07/07/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Organic anion transporting polypeptides (OATPs) are membrane proteins that mediate the uptake of a wide range of substrates across the plasma membrane of various cells and tissues. They are classified into 6 subfamilies, OATP1 through OATP6. Humans contain 12 OATPs encoded by 11 solute carrier of organic anion transporting polypeptide (SLCO) genes: OATP1A2, OATP1B1, OATP1B3, the splice variant OATP1B3-1B7, OATP1C1, OATP2A1, OATP2B1, OATP3A1, OATP4A1, OATP4C1, OATP5A1, and OATP6A1. Most of these proteins are expressed in epithelial cells, where they mediate the uptake of structurally unrelated organic anions, cations, and even neutral compounds into the cytoplasm. The best-characterized members are OATP1B1 and OATP1B3, which have an important role in drug metabolism by mediating drug uptake into the liver and are involved in drug-drug interactions. In this review, we aimed to (1) provide a historical perspective on the identification of OATPs and their nomenclature and discuss their phylogenic relationships and molecular characteristics; (2) review the current knowledge of the broad substrate specificity and their role in drug disposition and drug-drug interactions, with a special emphasis on human hepatic OATPs; (3) summarize the different experimental systems that are used to study the function of OATPs and discuss their advantages and disadvantages; (4) review the available experimental 3-dimensional structures and examine how they can help elucidate the transport mechanisms of OATPs; and (5) finally, summarize the current knowledge of the regulation of OATP expression, discuss clinically important single-nucleotide polymorphisms, and outline challenges of physiologically based pharmacokinetic modeling and in vitro to in vivo extrapolation. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (OATPs) are a family of 12 uptake transporters in the solute carrier superfamily. Several members, particularly the liver-expressed OATP1B1 and OATP1B3, are important drug transporters. They mediate the uptake of several endobiotics and xenobiotics, including statins and numerous other drugs, into hepatocytes, and their inhibition by other drugs or reduced expression due to single-nucleotide polymorphisms can lead to adverse drug effects. Their recently solved 3-dimensional structures should help to elucidate their transport mechanisms and broad substrate specificities.
Collapse
Affiliation(s)
- Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruno Stieger
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| | - Kaspar P Locher
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
2
|
Sato T, Kawabata T, Kumondai M, Hayashi N, Komatsu H, Kikuchi Y, Onoguchi G, Sato Y, Nanatani K, Hiratsuka M, Maekawa M, Yamaguchi H, Abe T, Tomita H, Mano N. Effect of Organic Anion Transporting Polypeptide 1B1 on Plasma Concentration Dynamics of Clozapine in Patients with Treatment-Resistant Schizophrenia. Int J Mol Sci 2024; 25:13228. [PMID: 39684938 DOI: 10.3390/ijms252313228] [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: 11/20/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
The involvement of drug-metabolizing enzymes and transporters in plasma clozapine (CLZ) dynamics has not been well examined in Japanese patients with treatment-resistant schizophrenia (TRS). Therefore, this clinical study investigated the relationship between single nucleotide polymorphisms (SNPs) of various pharmacokinetic factors (drug-metabolizing enzymes and transporters) and dynamic changes in CLZ. Additionally, we aimed to determine whether CLZ acts as a substrate for pharmacokinetic factors using in vitro assays and molecular docking calculations. We found that 6 out of 10 patients with TRS and with multiple organic anion transporting polypeptide (OATP) variants (OATP1B1: *1b, *15; OATP1B3: 334T>G, 699G>A; and OATP2B1: *3, 935G>A, 601G>A, 76_84del) seemed to be highly exposed to CLZ and/or N-desmethyl CLZ. A CLZ uptake study using OATP-expressing HEK293 cells showed that CLZ was a substrate of OATP1B1 with Km and Vmax values of 38.9 µM and 2752 pmol/mg protein/10 min, respectively. The results of molecular docking calculations supported the differences in CLZ uptake among OATP molecules and the weak inhibitory effect of cyclosporine A, which is a strong inhibitor of OATPs, on CLZ uptake via OATP1B1. This is the first study to show that CLZ is an OATP1B1 substrate and that the presence of SNPs in OATPs potentially alters CLZ pharmacokinetic parameters.
Collapse
Affiliation(s)
- Toshihiro Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Takeshi Kawabata
- Graduate School of Information Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masaki Kumondai
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Nagomi Hayashi
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Hiroshi Komatsu
- Department of Psychiatry, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Yuki Kikuchi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Go Onoguchi
- Department of Psychiatry, Tohoku University Hospital, Sendai 980-8574, Japan
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Yu Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Kei Nanatani
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan
| | - Masahiro Hiratsuka
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Japan
| | - Hiroaki Yamaguchi
- Department of Pharmacy, Yamagata University Hospital, Yamagata 990-9585, Japan
- Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Takaaki Abe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Graduate School of Medicine, Tohoku University, Sendai 980-8574, Japan
- Division of Medical Science, Graduate School of Biomedical Engineering, Tohoku University, Sendai 980-8579, Japan
- Department of Clinical Biology and Hormonal Regulation, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Tohoku University Hospital, Sendai 980-8574, Japan
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai 980-8574, Japan
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| |
Collapse
|
3
|
Dombi Á, Kaci H, Valentová K, Bakos É, Özvegy-Laczka C, Poór M. Interaction of myricetin, ampelopsin (dihydromyricetin), and their sulfate metabolites with serum albumin, cytochrome P450 (CYP2C9, 2C19, and 3A4) enzymes, and organic anion-transporting polypeptides (OATP1B1 and OATP2B1). Pharmacol Res Perspect 2024; 12:e70021. [PMID: 39344282 PMCID: PMC11440035 DOI: 10.1002/prp2.70021] [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: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Myricetin (MYR) and ampelopsin (AMP, or dihydromyricetin) are flavonoid aglycones found in certain plants and dietary supplements. During the presystemic biotransformation of flavonoids, mainly sulfate and glucuronide derivatives are produced, which are the dominant metabolites in the circulation. In this study, we tested the interactions of MYR, myricetin-3'-O-sulfate (M3'S), AMP, and ampelopsin-4'-O-sulfate (A4'S) with human serum albumin (HSA), cytochrome P450 enzymes (CYPs), and organic anion-transporting polypeptides (OATPs) using in vitro models, including the recently developed method for measuring flavonoid levels in living cells. M3'S and MYR bound to albumin with high affinity, and they showed moderate displacing effects versus the Site I marker warfarin. MYR, M3'S, AMP, and A4'S exerted no or only minor inhibitory effects on CYP2C9, CYP2C19, and CYP3A4 enzymes. M3'S and MYR caused considerable inhibitory actions on OATP1B1 at low micromolar concentrations (IC50 = 1.7 and 6.4 μM, respectively), while even their nanomolar levels resulted in strong inhibitory effects on OATP2B1 (IC50 = 0.3 and 0.4 μM, respectively). In addition, M3'S proved to be a substrate of OATP1B1 and OATP2B1. These results suggest that MYR-containing dietary supplements may affect the OATP-mediated transport of certain drugs, and OATPs are involved in the tissue uptake of M3'S.
Collapse
Affiliation(s)
- Ágnes Dombi
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Hana Kaci
- Drug Resistance Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, HUN-REN, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Éva Bakos
- Drug Resistance Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, HUN-REN, Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Drug Resistance Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, HUN-REN, Budapest, Hungary
| | - Miklós Poór
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Molecular Medicine Research Group, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
4
|
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
|
5
|
Linfield RY, Nguyen NN, Laprade OH, Holodniy M, Chary A. An update on drug-drug interactions in older adults living with human immunodeficiency virus (HIV). Expert Rev Clin Pharmacol 2024; 17:589-614. [PMID: 38753455 PMCID: PMC11233252 DOI: 10.1080/17512433.2024.2350968] [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: 02/01/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION People with HIV are living longer due to advances in antiretroviral therapy. With improved life expectancy comes an increased lifetime risk of comorbid conditions - such as cardiovascular disease and cancer - and polypharmacy. Older adults, particularly those living with HIV, are more vulnerable to drug interactions and adverse effects, resulting in negative health outcomes. AREA COVERED Antiretrovirals are involved in many potential drug interactions with medications used to treat common comorbidities and geriatric conditions in an aging population of people with HIV. We review the mechanisms and management of significant drug-drug interactions involving antiretroviral medications and non-antiretroviral medications commonly used among older people living with HIV. The management of these interactions may require dose adjustments, medication switches to alternatives, enhanced monitoring, and considerations of patient- and disease-specific factors. EXPERT OPINION Clinicians managing comorbid conditions among older people with HIV must be particularly vigilant to side effect profiles, drug-drug interactions, pill burden, and cost when optimizing treatment. To support healthier aging among people living with HIV, there is a growing need for antiretroviral stewardship, multidisciplinary care models, and advances that promote insight into the correlations between an individual, their conditions, and their medications.
Collapse
Affiliation(s)
| | - Nancy N. Nguyen
- Department of Pharmacy, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Pharmacy Practice, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, USA
| | - Olivia H. Laprade
- Department of Pharmacy, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Pharmacy Practice, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, USA
| | - Mark Holodniy
- Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- National Public Health Program Office, Veterans Health Administration, Palo Alto, CA, USA
| | - Aarthi Chary
- Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- National Public Health Program Office, Veterans Health Administration, Palo Alto, CA, USA
| |
Collapse
|
6
|
Toshimoto K. Beyond the basics: A deep dive into parameter estimation for advanced PBPK and QSP models. Drug Metab Pharmacokinet 2024; 56:101011. [PMID: 38833901 DOI: 10.1016/j.dmpk.2024.101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/26/2024] [Accepted: 03/14/2024] [Indexed: 06/06/2024]
Abstract
Physiologically-based pharmacokinetic (PBPK) models and quantitative systems pharmacology (QSP) models have contributed to drug development strategies. The parameters of these models are commonly estimated by capturing observed values using the nonlinear least-squares method. Software packages for PBPK and QSP modeling provide a range of parameter estimation algorithms. To choose the most appropriate method, modelers need to understand the basic concept of each approach. This review provides a general introduction to the key points of parameter estimation with a focus on the PBPK and QSP models, and the respective parameter estimation algorithms. The latter part assesses the performance of five parameter estimation algorithms - the quasi-Newton method, Nelder-Mead method, genetic algorithm, particle swarm optimization, and Cluster Gauss-Newton method - using three examples of PBPK and QSP modeling. The assessment revealed that some parameter estimation results were significantly influenced by the initial values. Moreover, the choice of algorithms demonstrating good estimation results heavily depends on factors such as model structure and the parameters to be estimated. To obtain credible parameter estimation results, it is advisable to conduct multiple rounds of parameter estimation under different conditions, employing various estimation algorithms.
Collapse
Affiliation(s)
- Kota Toshimoto
- Systems Pharmacology, Non-Clinical Biomedical Science, Applied Research & Operations, Astellas Pharma Inc., Ibaraki, Japan.
| |
Collapse
|
7
|
Kaci H, Bakos É, Needs PW, Kroon PA, Valentová K, Poór M, Özvegy-Laczka C. The 2-aminoethyl diphenylborinate-based fluorescent method identifies quercetin and luteolin metabolites as substrates of Organic anion transporting polypeptides, OATP1B1 and OATP2B1. Eur J Pharm Sci 2024; 196:106740. [PMID: 38437885 DOI: 10.1016/j.ejps.2024.106740] [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: 12/13/2023] [Revised: 01/28/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
Organic anion transporting polypeptides (OATPs), OATP1B1 and OATP2B1 are membrane proteins mediating the cellular uptake of chemically diverse organic compounds. OATP1B1 is exclusively expressed in hepatocytes and plays a key role in hepatic detoxification. The ubiquitously expressed OATP2B1 promotes the intestinal absorption of orally administered drugs. Flavonoids are widely found in foods and beverages, and many of them can inhibit OATP function, resulting in food-drug interactions. In our previous work, we have shown that not only luteolin (LUT) and quercetin (Q), but also some of their metabolites can inhibit OATP1B1 and OATP2B1 activity. However, data about the potential direct transport of these flavonoids by OATPs have been incomplete. Hence, in the current study, we developed a simple, fluorescence-based method for the measurement of intracellular flavonoid levels. The method applies a cell-permeable small molecule (2-aminoethyl diphenylborinate, 2-APB), that, upon forming a complex with flavonoids, results in their fluorescence enhancement. This way the direct uptake of LUT and Q, and also their metabolites' could be investigated both by confocal microscopy and in a fluorescence plate reader in living cells. With this approach we identified quercetin-3'-O-sulfate, luteolin-3'-O-glucuronide, luteolin-7-O-glucuronide and luteolin-3'-O-sulfate as substrates of both OATP1B1 and OATP2B1. Our results highlight that OATP1B1 and OATP2B1 can be key participants in the transmembrane movement of LUT and Q conjugates with otherwise low cell permeability. In addition, the novel method developed in this study can be a good completion to existing fluorescence-based assays to investigate OATP function.
Collapse
Affiliation(s)
- Hana Kaci
- Institute of Molecular Life Sciences, RCNS, HUN-REN, H-1117 Budapest, Magyar tudósok krt. 2., Hungary; Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, 1117 Budapest Pázmány Péter sétány 1/C, Hungary
| | - Éva Bakos
- Institute of Molecular Life Sciences, RCNS, HUN-REN, H-1117 Budapest, Magyar tudósok krt. 2., Hungary
| | - Paul W Needs
- Food, Microbiome & Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk NR4 7UQ, UK
| | - Paul A Kroon
- Food, Microbiome & Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk NR4 7UQ, UK
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague CZ-142 00, Czech Republic
| | - Miklós Poór
- Molecular Medicine Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary; Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, Pécs H-7624, Hungary; Department of Laboratory Medicine, Medical School, University of Pécs, Ifjúság útja 13, Pécs H-7624, Hungary
| | - Csilla Özvegy-Laczka
- Institute of Molecular Life Sciences, RCNS, HUN-REN, H-1117 Budapest, Magyar tudósok krt. 2., Hungary.
| |
Collapse
|
8
|
Powell JT, Kayesh R, Ballesteros-Perez A, Alam K, Niyonshuti P, Soderblom EJ, Ding K, Xu C, Yue W. Assessing Trans-Inhibition of OATP1B1 and OATP1B3 by Calcineurin and/or PPIase Inhibitors and Global Identification of OATP1B1/3-Associated Proteins. Pharmaceutics 2023; 16:63. [PMID: 38258074 PMCID: PMC10818623 DOI: 10.3390/pharmaceutics16010063] [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: 09/09/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 are key determinants of drug-drug interactions (DDIs). Various drugs including the calcineurin inhibitor (CNI) cyclosporine A (CsA) exert preincubation-induced trans-inhibitory effects upon OATP1B1 and/or OATP1B3 (abbreviated as OATP1B1/3) by unknown mechanism(s). OATP1B1/3 are phosphoproteins; calcineurin, which dephosphorylates and regulates numerous phosphoproteins, has not previously been investigated in the context of preincubation-induced trans-inhibition of OATP1B1/3. Herein, we compare the trans-inhibitory effects exerted on OATP1B1 and OATP1B3 by CsA, the non-analogous CNI tacrolimus, and the non-CNI CsA analogue SCY-635 in transporter-overexpressing human embryonic kidney (HEK) 293 stable cell lines. Preincubation (10-60 min) with tacrolimus (1-10 µM) rapidly and significantly reduces OATP1B1- and OATP1B3-mediated transport up to 0.18 ± 0.03- and 0.20 ± 0.02-fold compared to the control, respectively. Both CsA and SCY-635 can trans-inhibit OATP1B1, with the inhibitory effects progressively increasing over a 60 min preincubation time. At each equivalent preincubation time, CsA has greater trans-inhibitory effects toward OATP1B1 than SCY-635. Preincubation with SCY-635 for 60 min yielded IC50 of 2.2 ± 1.4 µM against OATP1B1, which is ~18 fold greater than that of CsA (0.12 ± 0.04 µM). Furthermore, a proteomics-based screening for protein interactors was used to examine possible proteins and processes contributing to OATP1B1/3 regulation and preincubation-induced inhibition by CNIs and other drugs. A total of 861 and 357 proteins were identified as specifically associated with OATP1B1 and OATP1B3, respectively, including various protein kinases, ubiquitin-related enzymes, the tacrolimus (FK506)-binding proteins FKBP5 and FKBP8, and several known regulatory targets of calcineurin. The current study reports several novel findings that expand our understanding of impaired OATP1B1/3 function; these include preincubation-induced trans-inhibition of OATP1B1/3 by the CNI tacrolimus, greater preincubation-induced inhibition by CsA compared to its non-CNI analogue SCY-635, and association of OATP1B1/3 with various proteins relevant to established and candidate OATP1B1/3 regulatory processes.
Collapse
Affiliation(s)
- John T. Powell
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Alexandra Ballesteros-Perez
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Khondoker Alam
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Pascaline Niyonshuti
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Erik J. Soderblom
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC 27708, USA
| | - Kai Ding
- Department of Biostatistics & Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.D.); (C.X.)
| | - Chao Xu
- Department of Biostatistics & Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.D.); (C.X.)
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| |
Collapse
|
9
|
Kikuchi R, Chothe PP, Chu X, Huth F, Ishida K, Ishiguro N, Jiang R, Shen H, Stahl SH, Varma MVS, Willemin ME, Morse BL. Utilization of OATP1B Biomarker Coproporphyrin-I to Guide Drug-Drug Interaction Risk Assessment: Evaluation by the Pharmaceutical Industry. Clin Pharmacol Ther 2023; 114:1170-1183. [PMID: 37750401 DOI: 10.1002/cpt.3062] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/08/2023] [Indexed: 09/27/2023]
Abstract
Drug-drug interactions (DDIs) involving hepatic organic anion transporting polypeptides 1B1/1B3 (OATP1B) can be substantial, however, challenges remain for predicting interaction risk. Emerging evidence suggests that endogenous biomarkers, particularly coproporphyrin-I (CP-I), can be used to assess in vivo OATP1B activity. The present work under the International Consortium for Innovation and Quality in Pharmaceutical Development was aimed primarily at assessing CP-I as a biomarker for informing OATP1B DDI risk. Literature and unpublished CP-I data along with pertinent in vitro and clinical DDI information were collected to identify DDIs primarily involving OATP1B inhibition and assess the relationship between OATP1B substrate drug and CP-I exposure changes. Static models to predict changes in exposure of CP-I, as a selective OATP1B substrate, were also evaluated. Significant correlations were observed between CP-I area under the curve ratio (AUCR) or maximum concentration ratio (Cmax R) and AUCR of substrate drugs. In general, the CP-I Cmax R was equal to or greater than the CP-I AUCR. CP-I Cmax R < 1.25 was associated with absence of OATP1B-mediated DDIs (AUCR < 1.25) with no false negative predictions. CP-I Cmax R < 2 was associated with weak OATP1B-mediated DDIs (AUCR < 2). A correlation was identified between CP-I exposure changes and OATP1B1 static DDI predictions. Recommendations for collecting and interpreting CP-I data are discussed, including a decision tree for guiding DDI risk assessment. In conclusion, measurement of CP-I is recommended to inform OATP1B inhibition potential. The current analysis identified changes in CP-I exposure that may be used to prioritize, delay, or replace clinical DDI studies.
Collapse
Affiliation(s)
- Ryota Kikuchi
- Quantitative, Translational and ADME Sciences, AbbVie Inc., North Chicago, Illinois, USA
| | - Paresh P Chothe
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Xiaoyan Chu
- ADME and Discovery Toxicology, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Felix Huth
- PK Sciences, Novartis Pharma AG, Basel, Switzerland
| | - Kazuya Ishida
- Drug Metabolism, Gilead Sciences Inc., Foster City, California, USA
| | - Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan
| | - Rongrong Jiang
- Drug Metabolism and Pharmacokinetics, Eisai Inc., Cambridge, Massachusetts, USA
| | - Hong Shen
- Departments of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, New Jersey, USA
| | - Simone H Stahl
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Manthena V S Varma
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut, USA
| | - Marie-Emilie Willemin
- Drug Metabolism and Pharmacokinetics, Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Bridget L Morse
- Department of Drug Disposition, Eli Lilly, Indianapolis, Indiana, USA
| |
Collapse
|
10
|
Mitra P, Kasliwala R, Iboki L, Madari S, Williams Z, Takahashi R, Taub ME. Mechanistic Static Model based Prediction of Transporter Substrate Drug-Drug Interactions Utilizing Atorvastatin and Rifampicin. Pharm Res 2023; 40:3025-3042. [PMID: 37821766 DOI: 10.1007/s11095-023-03613-x] [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: 06/30/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
OBJECTIVE An in vitro relative activity factor (RAF) technique combined with mechanistic static modeling was examined to predict drug-drug interaction (DDI) magnitude and analyze contributions of different clearance pathways in complex DDIs involving transporter substrates. Atorvastatin and rifampicin were used as a model substrate and inhibitor pair. METHODS In vitro studies were conducted with transfected HEK293 cells, hepatocytes and human liver microsomes. Prediction success was defined as predictions being within twofold of observations. RESULTS The RAF method successfully translated atorvastatin uptake from transfected cells to hepatocytes, demonstrating its ability to quantify transporter contributions to uptake. Successful translation of atorvastatin's in vivo intrinsic hepatic clearance (CLint,h,in vivo) from hepatocytes to liver was only achieved through consideration of albumin facilitated uptake or through application of empirical scaling factors to transporter-mediated clearances. Transporter protein expression differences between hepatocytes and liver did not affect CLint,h,in vivo predictions. By integrating cis and trans inhibition of OATP1B1/OATP1B3, atorvastatin-rifampicin (single dose) DDI magnitude could be accurately predicted (predictions within 0.77-1.0 fold of observations). Simulations indicated that concurrent inhibition of both OATP1B1 and OATP1B3 caused approximately 80% of atorvastatin exposure increases (AUCR) in the presence of rifampicin. Inhibiting biliary elimination, hepatic metabolism, OATP2B1, NTCP, and basolateral efflux are predicted to have minimal to no effect on AUCR. CONCLUSIONS This study demonstrates the effective application of a RAF-based translation method combined with mechanistic static modeling for transporter substrate DDI predictions and subsequent mechanistic interpretation.
Collapse
Affiliation(s)
- Pallabi Mitra
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Old Ridgebury Road, Ridgefield, CT, 06877, USA.
| | - Rumanah Kasliwala
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Laeticia Iboki
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Shilpa Madari
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Zachary Williams
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Ryo Takahashi
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Hyogo, Japan
| | - Mitchell E Taub
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| |
Collapse
|
11
|
Nozaki Y, Izumi S. Preincubation Time-Dependent, Long-Lasting Inhibition of Drug Transporters and Impact on the Prediction of Drug-Drug Interactions. Drug Metab Dispos 2023; 51:1077-1088. [PMID: 36854606 DOI: 10.1124/dmd.122.000970] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/05/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Transporter-mediated drug-drug interaction (DDI) is of clinical concern, and the quantitative prediction of DDIs is an indispensable part of drug development. Cell-based inhibition assays, in which a representative probe substrate and a potential inhibitor are coincubated, are routinely performed to assess the inhibitory potential of new molecular entities on drug transporters. However, the inhibitory effect of cyclosporine A (CsA) on organic anion transporting polypeptide (OATP) 1B1 is substantially potentiated with CsA preincubation, and this effect is both long-lasting and dependent on the preincubation time. This phenomenon has also been reported with transporters other than OATP1Bs, but it is considered more prevalent among OATP1Bs and organic cation transporters. Regulatory agencies have also noted this preincubation effect and have recommended that pharmaceutical companies consider inhibitor preincubation when performing in vitro OATP1B1 and OATP1B3 inhibition studies. Although the underlying mechanisms responsible for the preincubation effect are not fully understood, a trans-inhibition mechanism was recently demonstrated for OATP1B1 inhibition by CsA, in which CsA inhibited OATP1B1 not only extracellularly (cis-inhibition) but also intracellularly (trans-inhibition). Furthermore, the trans-inhibition potency of CsA was much greater than that of cis-inhibition, suggesting that trans-inhibition might be a key driver of clinical DDIs of CsA with OATP1B substrate drugs. Although confidence in transporter-mediated DDI prediction is generally considered to be low, the predictability might be further improved by incorporating the trans-inhibition mechanism into static and dynamic models for preincubation-dependent inhibitors of OATP1Bs and perhaps other transporters. SIGNIFICANCE STATEMENT: Preincubation time-dependent, long-lasting inhibition has been observed for OATP1B1 and other solute carrier transporters in vitro. Recently, a trans-inhibition mechanism for the preincubation effect of CsA on OATP1B1 inhibition was identified, with the trans-inhibition potency being greater than that of cis-inhibition. The concept of trans-inhibition may allow us to further understand the mechanism of transporter-mediated DDIs not only for OATP1B1 but also for other transporters and to improve the accuracy and confidence of DDI predictions.
Collapse
Affiliation(s)
- Yoshitane Nozaki
- Global Drug Metabolism and Pharmacokinetics, Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba, Ibaraki, 300-2635, Japan (Y.N., S.I.)
| | - Saki Izumi
- Global Drug Metabolism and Pharmacokinetics, Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba, Ibaraki, 300-2635, Japan (Y.N., S.I.)
| |
Collapse
|
12
|
Li Y, Drabison T, Nepal M, Ho RH, Leblanc AF, Gibson AA, Jin Y, Yang W, Huang KM, Uddin ME, Chen M, DiGiacomo DF, Chen X, Razzaq S, Tonniges JR, McTigue DM, Mims AS, Lustberg MB, Wang Y, Hummon AB, Evans WE, Baker SD, Cavaletti G, Sparreboom A, Hu S. Targeting a xenobiotic transporter to ameliorate vincristine-induced sensory neuropathy. JCI Insight 2023; 8:e164646. [PMID: 37347545 PMCID: PMC10443802 DOI: 10.1172/jci.insight.164646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 06/15/2023] [Indexed: 06/24/2023] Open
Abstract
Vincristine is a widely used chemotherapeutic drug for the treatment of multiple malignant diseases that causes a dose-limiting peripheral neurotoxicity. There is no clinically effective preventative treatment for vincristine-induced sensory peripheral neurotoxicity (VIPN), and mechanistic details of this side effect remain poorly understood. We hypothesized that VIPN is dependent on transporter-mediated vincristine accumulation in dorsal root ganglion neurons. Using a xenobiotic transporter screen, we identified OATP1B3 as a neuronal transporter regulating the uptake of vincristine. In addition, genetic or pharmacological inhibition of the murine orthologue transporter OATP1B2 protected mice from various hallmarks of VIPN - including mechanical allodynia, thermal hyperalgesia, and changes in digital maximal action potential amplitudes and neuronal morphology - without negatively affecting plasma levels or antitumor effects of vincristine. Finally, we identified α-tocopherol from an untargeted metabolomics analysis as a circulating endogenous biomarker of neuronal OATP1B2 function, and it could serve as a companion diagnostic to guide dose selection of OATP1B-type transport modulators given in combination with vincristine to prevent VIPN. Collectively, our findings shed light on the fundamental basis of VIPN and provide a rationale for the clinical development of transporter inhibitors to prevent this debilitating side effect.
Collapse
Affiliation(s)
- Yang Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
- Division of Outcomes and Translational Sciences, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Thomas Drabison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Mahesh Nepal
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
- Division of Outcomes and Translational Sciences, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Richard H. Ho
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alix F. Leblanc
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Alice A. Gibson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Yan Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kevin M. Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Muhammad Erfan Uddin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Mingqing Chen
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Duncan F. DiGiacomo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Xihui Chen
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Sobia Razzaq
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | | | - Dana M. McTigue
- The Belford Center for Spinal Cord Injury & Department of Neuroscience, College of Medicine, and
| | - Alice S. Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Maryam B. Lustberg
- The Breast Center at Smilow Cancer Hospital at Yale, New Haven, Connecticut, USA
| | - Yijia Wang
- Department of Chemistry and Biochemistry & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Amanda B. Hummon
- Department of Chemistry and Biochemistry & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - William E. Evans
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Guido Cavaletti
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, and
- Division of Outcomes and Translational Sciences, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
13
|
Schulz JA, Stresser DM, Kalvass JC. Plasma Protein-Mediated Uptake and Contradictions to the Free Drug Hypothesis: A Critical Review. Drug Metab Rev 2023:1-34. [PMID: 36971325 DOI: 10.1080/03602532.2023.2195133] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
According to the free drug hypothesis (FDH), only free, unbound drug is available to interact with biological targets. This hypothesis is the fundamental principle that continues to explain the vast majority of all pharmacokinetic and pharmacodynamic processes. Under the FDH, the free drug concentration at the target site is considered the driver of pharmacodynamic activity and pharmacokinetic processes. However, deviations from the FDH are observed in hepatic uptake and clearance predictions, where observed unbound intrinsic hepatic clearance (CLint,u) is larger than expected. Such deviations are commonly observed when plasma proteins are present and form the basis of the so-called plasma protein-mediated uptake effect (PMUE). This review will discuss the basis of plasma protein binding as it pertains to hepatic clearance based on the FDH, as well as several hypotheses that may explain the underlying mechanisms of PMUE. Notably, some, but not all, potential mechanisms remained aligned with the FDH. Finally, we will outline possible experimental strategies to elucidate PMUE mechanisms. Understanding the mechanisms of PMUE and its potential contribution to clearance underprediction is vital to improving the drug development process.
Collapse
|
14
|
Kayesh R, Tambe V, Xu C, Yue W. Differential Preincubation Effects of Nicardipine on OATP1B1- and OATP1B3-Mediated Transport in the Presence and Absence of Protein: Implications in Assessing OATP1B1- and OATP1B3-Mediated Drug-Drug Interactions. Pharmaceutics 2023; 15:1020. [PMID: 36986880 PMCID: PMC10052025 DOI: 10.3390/pharmaceutics15031020] [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: 12/10/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Impaired transport activity of hepatic OATP1B1 and OATP1B3 due to drug-drug interactions (DDIs) often leads to increased systemic exposure to substrate drugs (e.g., lipid-lowering statins). Since dyslipidemia and hypertension frequently coexist, statins are often concurrently used with antihypertensives, including calcium channel blockers (CCBs). OATP1B1/1B3-related DDIs in humans have been reported for several CCBs. To date, the OATP1B1/1B3-mediated DDI potential of CCB nicardipine has not been assessed. The current study was designed to assess the OATP1B1- and OATP1B3-mediated DDI potential of nicardipine using the R-value model, following the US-FDA guidance. IC50 values of nicardipine against OATP1B1 and OATP1B3 were determined in transporter-overexpressing human embryonic kidney 293 cells using [3H]-estradiol 17β-D-glucuronide and [3H]-cholecystokinin-8 as substrates, respectively, with or without nicardipine-preincubation in protein-free Hanks' Balanced Salt Solution (HBSS) or in fetal bovine serum (FBS)-containing culture medium. Preincubation with nicardipine for 30 min in protein-free HBSS buffer produced lower IC50 and higher R-values for both OATP1B1 and OATP1B3 compared to in FBS-containing medium, yielding IC50 values of 0.98 and 1.63 µM and R-values of 1.4 and 1.3 for OATP1B1 and OATP1B3, respectively. The R-values were higher than the US-FDA cut-off value of 1.1, supporting that nicardipine has the potential to cause OATP1B1/3-mediated DDIs. Current studies provide insight into the consideration of optimal preincubation conditions when assessing the OATP1B1/3-mediated DDIs in vitro.
Collapse
Affiliation(s)
- Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Vishakha Tambe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| |
Collapse
|
15
|
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
|
16
|
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]
|
17
|
Martin P, Czerwiński M, Limaye PB, Ogilvie BW, Smith S, Boyd B. In vitro evaluation suggests fenfluramine and norfenfluramine are unlikely to act as perpetrators of drug interactions. Pharmacol Res Perspect 2022; 10:e00959. [PMID: 35599347 PMCID: PMC9124818 DOI: 10.1002/prp2.959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
Studies support the safety and efficacy of fenfluramine (FFA) as an antiseizure medication (ASM) in Dravet syndrome, Lennox-Gastaut syndrome, or CDKL5 deficiency disorder, all pharmacoresistant developmental and epileptic encephalopathies. However, drug-drug interactions with FFA in multi-ASM regimens have not been fully investigated. We characterized the perpetrator potential of FFA and its active metabolite, norfenfluramine (nFFA), in vitro by assessing cytochrome P450 (CYP450) inhibition in human liver microsomes, CYP450 induction in cultured human hepatocytes, and drug transporter inhibition potential in permeability or cellular uptake assays. Mean plasma unbound fraction was ~50% for both FFA and nFFA, with no apparent concentration dependence. FFA and nFFA were direct in vitro inhibitors of CYP2D6 (IC50 , 4.7 and 16 µM, respectively) but did not substantially inhibit CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, or CYP3A4/5. No time- or metabolism-dependent CYP450 inhibition occurred. FFA and nFFA did not induce CYP1A2; both induced CYP2B6 (up to 2.8-fold and up to 2.0-fold, respectively) and CYP3A4 (1.9- to 3.0-fold and 3.6- to 4.8-fold, respectively). Mechanistic static pharmacokinetic models predicted that neither CYP450 inhibition nor induction was likely to be clinically relevant at doses typically used for seizure reduction (ratio of area under curve [AUCR] for inhibition <1.25; AUCR for induction >0.8). Transporters OCT2 and MATE1 were inhibited by FFA (IC50 , 19.8 and 9.0 μM) and nFFA (IC50 , 5.2 and 4.6 μM) at concentrations higher than clinically achievable; remaining transporters were not inhibited. Results suggest that FFA and nFFA are unlikely drug-drug interaction perpetrators at clinically relevant doses of FFA (0.2-0.7 mg/kg/day).
Collapse
|
18
|
Martin P, Czerwiński M, Limaye PB, Muranjan S, Ogilvie BW, Smith S, Boyd B. In vitro evaluation of fenfluramine and norfenfluramine as victims of drug interactions. Pharmacol Res Perspect 2022; 10:e00958. [PMID: 35599345 PMCID: PMC9124820 DOI: 10.1002/prp2.958] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/06/2022] [Indexed: 12/27/2022] Open
Abstract
Fenfluramine (FFA) has potent antiseizure activity in severe, pharmacoresistant childhood‐onset developmental and epileptic encephalopathies (e.g., Dravet syndrome). To assess risk of drug interaction affecting pharmacokinetics of FFA and its major metabolite, norfenfluramine (nFFA), we conducted in vitro metabolite characterization, reaction phenotyping, and drug transporter−mediated cellular uptake studies. FFA showed low in vitro clearance in human liver S9 fractions and in intestinal S9 fractions in all three species tested (t1/2 > 120 min). Two metabolites (nFFA and an N‐oxide or a hydroxylamine) were detected in human liver microsomes versus six in dog and seven in rat liver microsomes; no metabolite was unique to humans. Selective CYP inhibitor studies showed FFA metabolism partially inhibited by quinidine (CYP2D6, 48%), phencyclidine (CYP2B6, 42%), and furafylline (CYP1A2, 32%) and, to a lesser extent (<15%), by tienilic acid (CYP2C9), esomeprazole (CYP2C19), and troleandomycin (CYP3A4/5). Incubation of nFFA with rCYP1A2, rCYP2B6, rCYP2C19, and rCYP2D6 resulted in 10%−20% metabolism and no clear inhibition of nFFA metabolism by any CYP‐selective inhibitor. Reaction phenotyping showed metabolism of FFA by recombinant human cytochrome P450 (rCYP) enzymes rCYP2B6 (10%–21% disappearance for 1 and 10 µM FFA, respectively), rCYP1A2 (22%−23%), rCYP2C19 (49%−50%), and rCYP2D6 (59%−97%). Neither FFA nor nFFA was a drug transporter substrate. Results show FFA metabolism to nFFA occurs through multiple pathways of elimination. FFA dose adjustments may be needed when administered with strong inhibitors or inducers of multiple enzymes involved in FFA metabolism (e.g., stiripentol).
Collapse
|
19
|
Izumi S, Nozaki Y, Lee W, Sugiyama Y. Experimental and Modeling Evidence Supporting the Trans-Inhibition Mechanism for Preincubation Time-Dependent, Long-Lasting Inhibition of Organic Anion Transporting Polypeptide 1B1 by Cyclosporine A. Drug Metab Dispos 2022; 50:541-551. [PMID: 35241487 DOI: 10.1124/dmd.121.000783] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/14/2022] [Indexed: 11/22/2022] Open
Abstract
Cyclosporine A (CsA) and rifampin are potent inhibitors of organic anion transporting polypeptide (OATP) 1B1 and are widely used to assess the risk for drug-drug interactions. CsA displays preincubation time-dependent, long-lasting inhibition of OATP1B1 in vitro and in rats in vivo, and a proposed mechanism is the trans-inhibition by which CsA inhibits OATP1B1 from the inside of cells. The current study aimed to experimentally validate the proposed mechanism using human embryonic kidney 293 cells stably expressing OATP1B1. The uptake of CsA reached a plateau following an approximate 60-minute incubation, with the cell-to-buffer concentration ratio of 3930, reflective of the high-affinity, high-capacity intracellular binding of CsA. The time course of CsA uptake was analyzed to estimate the kinetic parameters for permeability clearance and intracellular binding. When the OATP1B1-mediated uptake of [3H]estradiol-17β-glucuronide was measured following preincubation with CsA for 5 to 120 minutes, apparent Ki values became lower with longer preincubation. Our kinetic modeling incorporated the two reversible inhibition constants [Ki,trans and Ki,cis for the inhibition from inside (trans-inhibition) and outside (cis-inhibition) of cells, respectively] and estimated Ki,trans value of CsA was smaller by 48-fold than the estimated Ki,cis value. Rifampin also displayed preincubation time-dependent inhibition of OATP1B1, albeit the extent of enhancement was only twofold. The current study provides experimental evidence for the preincubation time-dependent shift of apparent Ki values and a mechanistic basis for physiologically based pharmacokinetic modeling that incorporates permeability clearance, extensive intracellular binding, and asymmetry of Ki values between the inside and outside of cells. SIGNIFICANCE STATEMENT: In vitro data and kinetic modeling support that preincubation time-dependent, long-lasting inhibition of OATP1B1 by CsA can be explained by the extensive intracellular binding and reversible OATP1B1 inhibition intracellularly (trans-inhibition) as well as extracellularly (cis-inhibition). For inhibitors to display time-dependency, the following factors were found important: time to reach a steady-state cellular concentration, trans-inhibition potency relative to cis-inhibition, and the degree of cellular inhibitor accumulation. This study would aid in the accurate prediction of drug-drug interactions mediated by OATP1B1 inhibition.
Collapse
Affiliation(s)
- Saki Izumi
- Global Drug Metabolism and Pharmacokinetics, Tsukuba Research Laboratories, Eisai Co. Ltd., Tsukuba, Japan (S.I., Y.N.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.L.); and Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Tokyo, Japan (Y.S.)
| | - Yoshitane Nozaki
- Global Drug Metabolism and Pharmacokinetics, Tsukuba Research Laboratories, Eisai Co. Ltd., Tsukuba, Japan (S.I., Y.N.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.L.); and Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Tokyo, Japan (Y.S.)
| | - Wooin Lee
- Global Drug Metabolism and Pharmacokinetics, Tsukuba Research Laboratories, Eisai Co. Ltd., Tsukuba, Japan (S.I., Y.N.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.L.); and Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Tokyo, Japan (Y.S.)
| | - Yuichi Sugiyama
- Global Drug Metabolism and Pharmacokinetics, Tsukuba Research Laboratories, Eisai Co. Ltd., Tsukuba, Japan (S.I., Y.N.); College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.L.); and Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Tokyo, Japan (Y.S.)
| |
Collapse
|
20
|
Sakurai K, Kuroda T, Abe J, Toda H, Kitamoto S. Identification of the organic anion transporting polypeptides responsible for the hepatic uptake of the major metabolite of epyrifenacil, S-3100-CA, in mice. Pharmacol Res Perspect 2021; 9:e00877. [PMID: 34619012 PMCID: PMC8496750 DOI: 10.1002/prp2.877] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/07/2021] [Indexed: 01/06/2023] Open
Abstract
Epyrifenacil is a novel herbicide that acts as an inhibitor of protoporphyrinogen oxidase (PPO) and produces hepatotoxicity in rodents by inhibiting PPO. Our previous research revealed that the causal substance of hepatotoxicity is S-3100-CA, a major metabolite of epyrifenacil, and that human hepatocyte uptake of S-3100-CA was significantly lower than rodent one, suggesting less relevant to hepatotoxicity in humans. To clarify the species difference in the uptake of S-3100-CA, we focused on organic anion transporting polypeptides (OATPs) and carried out an uptake assay using human, rat, and mouse OATP hepatic isoforms-expressing 293FT cells. As a result, all the examined OATPs were found to contribute to the S-3100-CA uptake, suggesting that the species difference was not due to the differences in selectivity toward OATP isoforms. When [14 C]epyrifenacil was administered to mice, the liver concentration of S-3100-CA was higher in males than in females. Furthermore, when [14 C]epyrifenacil was administered with OATP inhibitors, the liver/plasma ratio of S-3100-CA was significantly decreased by rifampicin, an Oatp1a1/Oatp1a4 inhibitor in mice, but not by digoxin, an Oatp1a4-specific inhibitor. This result indicates that Oatp1a1, the predominant transporter in male mice, is the main contributor to the hepatic transport of S-3100-CA, and consequently to the gender difference. Moreover, we conclude that the species difference in the hepatic uptake of S-3100-CA observed in our previous research is not due to differences in the selectivity toward OATP isoforms but rather to the significantly higher expression of OATPs which mediate uptake of S-3100-CA in rodents than in humans.
Collapse
Affiliation(s)
- Kengo Sakurai
- Environmental Health Science LaboratorySumitomo Chemical Co., Ltd.OsakaJapan
| | - Tomohiro Kuroda
- Environmental Health Science LaboratorySumitomo Chemical Co., Ltd.OsakaJapan
| | - Jun Abe
- Environmental Health Science LaboratorySumitomo Chemical Co., Ltd.OsakaJapan
| | - Hiroshi Toda
- Environmental Health Science LaboratorySumitomo Chemical Co., Ltd.OsakaJapan
| | - Sachiko Kitamoto
- Environmental Health Science LaboratorySumitomo Chemical Co., Ltd.OsakaJapan
| |
Collapse
|
21
|
Ungvári O, Király L, Bakos É, Özvegy-Laczka C. 8-acetoxy-trisulfopyrene as the first activatable fluorogenic probe for add-and-read assessment of Organic anion-transporting polypeptides, OATP1B1, OATP1B3, and OATP2B1. FASEB J 2021; 35:e21863. [PMID: 34411334 DOI: 10.1096/fj.202100648r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/15/2021] [Accepted: 08/05/2021] [Indexed: 12/30/2022]
Abstract
Organic anion-transporting polypeptides, OATP1B1, OATP1B3, and OATP2B1 are multispecific membrane proteins mediating the hepatocellular uptake of structurally diverse endo- and exogenous compounds, including various kinds of drugs. Co-administration of OATP1B/2B1 substrates may lead to altered pharmacokinetics or even toxicity. Therefore, the study of the interaction with these OATPs is essential in drug development and is recommended by international regulatory agencies, the FDA, EMA, and PMDA. In general, radiolabeled indicators are used to measure drug interactions of OATPs, and, lately, fluorescent probes are also gaining wider application in OATP tests. However, all of the currently available methods (either radioactive or fluorescence-based) comprise multiple steps, including the removal of the indicator in the end of the experiment. Hence, they are not ideally suited for high-throughput screening. In the current study, in order to find an indicator allowing real-time assessment of hepatic OATP function, we searched for an activatable fluorogenic OATP substrate. Here, we show that 8-acetoxypyrene-1,3,6-trisulfonate (Ace), a fluorogenic derivative of the hepatic OATP substrate pyranine (8-hydroxypyrene-1,3,6-trisulfonate) enters the cells via OATP1B1/3 or OATP2B1 function. In living cells, Ace is then converted into highly fluorescent pyranine, allowing "no-wash" measurement of OATP function and drug interactions. Furthermore, we demonstrate that Ace can be used in an indirect assay termed as competitive counterflow suitable to distinguish between transported substrates and inhibitors of OATP1B1. The fluorescence-based methods described here are unique and open the way toward high-throughput screening of interactions between new molecular entities and OATPs.
Collapse
Affiliation(s)
- Orsolya Ungvári
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| | - Laura Király
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| | - Éva Bakos
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| |
Collapse
|
22
|
Fashe M, Yi M, Sueyoshi T, Negishi M. Sex-specific expression mechanism of hepatic estrogen inactivating enzyme and transporters in diabetic women. Biochem Pharmacol 2021; 190:114662. [PMID: 34157297 DOI: 10.1016/j.bcp.2021.114662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 12/21/2022]
Abstract
Circulating estrogens levels significantly decrease in menopause and levels off in postmenopausal women. Accordingly, the liver represses levels of enzymes and membrane transporters, thereby decreasing capability of inactivating and excreting estrogens. Women increasingly develop type 2 diabetes during or after menopause. Estrogens are known to promote liver diseases in these women. Here, we have found that the estrogen inactivating sulfotransferase (SULT1E1) and an ATP-binding cassette subfamily G member 2 (ABCG2), a gene encoding breast cancer resistance protein that exports sulfated estrogens, increased their expression levels in diabetic women but not men. For the sulfotransferase gene, phosphorylated nuclear receptors ERα and RORα, at Ser212 and Ser100, respectively, bind their response elements to activate the SULT1E1 promoter in women. This coordinated increase in estrogen inactivation and excretion, and the phosphorylated nuclear receptor-mediated gene activation could be a defense mechanism against toxicities of estrogens through inactivation and excretion in the livers of women.
Collapse
Affiliation(s)
- Muluneh Fashe
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | - MyeongJin Yi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Tatsuya Sueyoshi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Masahiko Negishi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
23
|
Tabernilla A, dos Santos Rodrigues B, Pieters A, Caufriez A, Leroy K, Van Campenhout R, Cooreman A, Gomes AR, Arnesdotter E, Gijbels E, Vinken M. In Vitro Liver Toxicity Testing of Chemicals: A Pragmatic Approach. Int J Mol Sci 2021; 22:5038. [PMID: 34068678 PMCID: PMC8126138 DOI: 10.3390/ijms22095038] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
The liver is among the most frequently targeted organs by noxious chemicals of diverse nature. Liver toxicity testing using laboratory animals not only raises serious ethical questions, but is also rather poorly predictive of human safety towards chemicals. Increasing attention is, therefore, being paid to the development of non-animal and human-based testing schemes, which rely to a great extent on in vitro methodology. The present paper proposes a rationalized tiered in vitro testing strategy to detect liver toxicity triggered by chemicals, in which the first tier is focused on assessing general cytotoxicity, while the second tier is aimed at identifying liver-specific toxicity as such. A state-of-the-art overview is provided of the most commonly used in vitro assays that can be used in both tiers. Advantages and disadvantages of each assay as well as overall practical considerations are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (A.T.); (B.d.S.R.); (A.P.); (A.C.); (K.L.); (R.V.C.); (A.C.); (A.R.G.); (E.A.); (E.G.)
| |
Collapse
|
24
|
Yee SW, Vora B, Oskotsky T, Zou L, Jakobsen S, Enogieru OJ, Koleske ML, Kosti I, Rödin M, Sirota M, Giacomini KM. Drugs in COVID-19 Clinical Trials: Predicting Transporter-Mediated Drug-Drug Interactions Using In Vitro Assays and Real-World Data. Clin Pharmacol Ther 2021; 110:108-122. [PMID: 33759449 PMCID: PMC8217266 DOI: 10.1002/cpt.2236] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/14/2021] [Indexed: 12/25/2022]
Abstract
Numerous drugs are currently under accelerated clinical investigation for the treatment of coronavirus disease 2019 (COVID‐19); however, well‐established safety and efficacy data for these drugs are limited. The goal of this study was to predict the potential of 25 small molecule drugs in clinical trials for COVID‐19 to cause clinically relevant drug‐drug interactions (DDIs), which could lead to potential adverse drug reactions (ADRs) with the use of concomitant medications. We focused on 11 transporters, which are targets for DDIs. In vitro potency studies in membrane vesicles or HEK293 cells expressing the transporters coupled with DDI risk assessment methods revealed that 20 of the 25 drugs met the criteria from regulatory authorities to trigger consideration of a DDI clinical trial. Analyses of real‐world data from electronic health records, including a database representing nearly 120,000 patients with COVID‐19, were consistent with several of the drugs causing transporter‐mediated DDIs (e.g., sildenafil, chloroquine, and hydroxychloroquine). This study suggests that patients with COVID‐19, who are often older and on various concomitant medications, should be carefully monitored for ADRs. Future clinical studies are needed to determine whether the drugs that are predicted to inhibit transporters at clinically relevant concentrations, actually result in DDIs.
Collapse
Affiliation(s)
- Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Bianca Vora
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California, USA
| | - Ling Zou
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Sebastian Jakobsen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Osatohanmwen J Enogieru
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Megan L Koleske
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Idit Kosti
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California, USA
| | - Mattias Rödin
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
25
|
Effects of Probenecid on Hepatic and Renal Disposition of Hexadecanedioate, an Endogenous Substrate of Organic Anion Transporting Polypeptide 1B in Rats. J Pharm Sci 2021; 110:2274-2284. [PMID: 33607188 DOI: 10.1016/j.xphs.2021.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 01/02/2023]
Abstract
The aim of the present study was to investigate changes in plasma concentrations and tissue distribution of endogenous substrates of organic anion transporting polypeptide (OATP) 1B, hexadecanedioate (HDA), octadecanedioate (ODA), tetradecanedioate (TDA), and coproporphyrin-III, induced by its weak inhibitor, probenecid (PBD), in rats. PBD increased the plasma concentrations of these four compounds regardless of bile duct cannulation, whereas liver-to-plasma (Kp,liver) and kidney-to-plasma concentration ratios of HDA and TDA were reduced. Similar effects of PBD on plasma concentrations and Kp,liver of HDA, ODA, and TDA were observed in kidney-ligated rats, suggesting a minor contribution of renal disposition to the overall distribution of these three compounds. Tissue uptake clearance of deuterium-labeled HDA (d-HDA) in liver was 16-fold higher than that in kidney, and was reduced by 80% by PBD. This was compatible with inhibition by PBD of d-HDA uptake in isolated rat hepatocytes. Such inhibitory effects of PBD were also observed in the human OATP1B1-mediated uptake of d-HDA. Overall, the disposition of HDA is mainly determined by hepatic OATP-mediated uptake, which is inhibited by PBD. HDA might, thus, be a biomarker for OATPs minimally affected by urinary and biliary elimination in rats.
Collapse
|
26
|
Bruyère A, Le Vée M, Jouan E, Molez S, Nies AT, Fardel O. Differential in vitro interactions of the Janus kinase inhibitor ruxolitinib with human SLC drug transporters. Xenobiotica 2021; 51:467-478. [PMID: 33455503 DOI: 10.1080/00498254.2021.1875516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Interactions of the Janus kinase (JAK) inhibitor ruxolitinib with solute carriers (SLCs) remain incompletely characterised. The present study was therefore designed to investigate this issue.The interactions of ruxolitinib with SLCs were analysed using transporter-overexpressing human embryonic kidney HEK293 cells. Substrate accumulation was detected by spectrofluorimetry, liquid chromatography coupled to tandem mass spectrometry or scintillation counting.Ruxolitinib was found to potently inhibit the activities of organic anion transporter 3 (OAT3), organic cation transporter 2 (OCT2), multidrug and toxin extrusion 1 (MATE1) and MATE2-K (half maximal inhibitory concentration (IC50) < 10 µM). It blocked OAT1, OAT4, OATP1B1, OATP1B3, OATP2B1 and OCT3, but in a weaker manner (IC50 > 10 µM), whereas OCT1 was not impacted. No time-dependent inhibition was highlighted. When applying the US Food and Drug Administration (FDA) criteria for transporters-related drug-drug interaction risk, OCT2 and MATE2-K, unlike MATE1 and OAT3, were predicted to be in vivo inhibited by ruxolitinib. Cellular uptake studies additionally indicated that ruxolitinib is a substrate for MATE1 and MATE2-K, but not for OAT3 and OCT2.Ruxolitinib in vitro blocked activities of most of SLC transporters. Only OCT2 and MATE-2K may be however clinically inhibited by the JAK inhibitor, with the caution for OCT2 that in vitro inhibition data were generated with an FDA-non recommended fluorescent substrate. Ruxolitinib MATEs-mediated transport may additionally deserve attention for its possible pharmacological consequences in MATE-positive cells.
Collapse
Affiliation(s)
- Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Marc Le Vée
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Elodie Jouan
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Stephanie Molez
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Anne T Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Stuttgart, Germany.,iFIT Cluster of Excellence (EXC2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Olivier Fardel
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
27
|
Kayesh R, Farasyn T, Crowe A, Liu Q, Pahwa S, Alam K, Neuhoff S, Hatley O, Ding K, Yue W. Assessing OATP1B1- and OATP1B3-Mediated Drug-Drug Interaction Potential of Vemurafenib Using R-Value and Physiologically-Based Pharmacokinetic Models. J Pharm Sci 2021; 110:314-324. [PMID: 32590030 PMCID: PMC7750294 DOI: 10.1016/j.xphs.2020.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 11/19/2022]
Abstract
Organic anion transporting polypeptides (OATP) 1B1 and OATP1B3 are important determinants of transporter-mediated drug-drug interactions (DDIs). Current studies assessed the OATP1B1 and OATP1B3-mediated DDI potential of vemurafenib, a kinase inhibitor drug with high protein binding and low aqueous solubility, using R-value and physiologically-based pharmacokinetic (PBPK) models. The total half-maximal inhibitory concentration (IC50,total) values of vemurafenib against OATP1B1 and OATP1B3 were determined in 100% human plasma in transporter-overexpressing human embryonic kidney 293 stable cell lines. The unbound fraction of vemurafenib in human plasma before (fu,plasma) and after addition into the uptake assay plate (fu,plasma,inc) were determined by rapid equilibrium dialysis. There was no statistically significant difference between fu,plasma and fu,plasma,inc. Vemurafenib IC50,total values against OATP1B1 and OATP1B3 are 175 ± 82 and 231 ± 26 μM, respectively. The R-values [R = 1 + fu,plasma × Iin,max/(fu,plasma,inc × IC50,total)] were then simplified as R = 1+Iin,max/IC50,total, and were 1.76 and 1.57 for OATP1B1 and OATP1B3, respectively. The simulated pravastatin AUC ratio was 1.28 when a single dose of pravastatin (40 mg) was co-administered with vemurafenib (960 mg, twice daily) at steady-state, compared to pravastatin alone. Both R-value and PBPK models predict that vemurafenib has the potential to cause OATP1B1- and OATP1B3-mediated DDIs.
Collapse
Affiliation(s)
- Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Taleah Farasyn
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Alexandra Crowe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Qiang Liu
- ARL Bio Pharma, Oklahoma City, Oklahoma 73104
| | - Sonia Pahwa
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Khondoker Alam
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Sibylle Neuhoff
- Certara UK Ltd, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ United Kingdom
| | - Oliver Hatley
- Certara UK Ltd, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ United Kingdom
| | - Kai Ding
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104.
| |
Collapse
|
28
|
Masuo Y, Fujita KI, Mishiro K, Seba N, Kogi T, Okumura H, Matsumoto N, Kunishima M, Kato Y. 6-Hydroxyindole is an endogenous long-lasting OATP1B1 inhibitor elevated in renal failure patients. Drug Metab Pharmacokinet 2020; 35:555-562. [PMID: 33191090 DOI: 10.1016/j.dmpk.2020.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/21/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022]
Abstract
The hepatic uptake transporter organic anion transporting polypeptide (OATP) 1B1 is inhibited by some uremic toxins; however, direct inhibition can only partially explain the delayed systemic elimination of substrate drugs in renal failure patients. This study aimed to examine the long-lasting inhibition of OATP1B1 by uremic toxins and their metabolites. Preincubation of HEK293/OATP1B1 cells with 21 uremic toxins resulted in almost no change in the uptake of a typical substrate [3H]estrone-3-sulfate (E1S), although some directly inhibited [3H]E1S uptake. In contrast, preincubation with an indole metabolite, 6-hydroxyindole, reduced [3H]E1S uptake, even after the inhibitor was washed out before [3H]E1S incubation. Such long-lasting inhibition by 6-hydroxyindole was time-dependent and recovered after a 3-h incubation without 6-hydroxyindole. Preincubation with 6-hydroxyindole increased the Km for [3H]E1S uptake with minimal change in Vmax. This was compatible with no change in the cell-surface expression of OATP1B1, as assessed by a biotinylation assay. Preincubation with 6-hydroxyindole reduced [3H]E1S uptake in human hepatocytes without changes in OATP1B1 mRNA. Plasma concentration of 6-hydroxyindole in renal failure patients increased as renal function decreased, but might be insufficient to exhibit potent OATP1B1 inhibition. In conclusion, 6-hydroxyindole is an endogenous long-lasting OATP1B1 inhibitor with elevated plasma concentrations in renal failure patients.
Collapse
Affiliation(s)
- Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ken-Ichi Fujita
- Division of Cancer Genome and Pharmacotherapy, Department of Clinical Pharmacy, Showa University School of Pharmacy, Tokyo, Japan
| | - Kenji Mishiro
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Natsumi Seba
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tatsuya Kogi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hidenori Okumura
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Natsumi Matsumoto
- Division of Cancer Genome and Pharmacotherapy, Department of Clinical Pharmacy, Showa University School of Pharmacy, Tokyo, Japan
| | - Munetaka Kunishima
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
29
|
Mohos V, Fliszár-Nyúl E, Ungvári O, Bakos É, Kuffa K, Bencsik T, Zsidó BZ, Hetényi C, Telbisz Á, Özvegy-Laczka C, Poór M. Effects of Chrysin and Its Major Conjugated Metabolites Chrysin-7-Sulfate and Chrysin-7-Glucuronide on Cytochrome P450 Enzymes and on OATP, P-gp, BCRP, and MRP2 Transporters. Drug Metab Dispos 2020; 48:1064-1073. [PMID: 32661014 DOI: 10.1124/dmd.120.000085] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022] Open
Abstract
Chrysin is an abundant flavonoid in nature, and it is also contained by several dietary supplements. Chrysin is highly biotransformed in the body, during which conjugated metabolites chrysin-7-sulfate and chrysin-7-glucuronide are formed. These conjugates appear at considerably higher concentrations in the circulation than the parent compound. Based on previous studies, chrysin can interact with biotransformation enzymes and transporters; however, the interactions of its metabolites have been barely examined. In this in vitro study, the effects of chrysin, chrysin-7-sulfate, and chrysin-7-glucuronide on cytochrome P450 enzymes (2C9, 2C19, 3A4, and 2D6) as well as on organic anion-transporting polypeptides (OATPs; 1A2, 1B1, 1B3, and 2B1) and ATP binding cassette [P-glycoprotein, multidrug resistance-associated protein 2, and breast cancer resistance protein (BCRP)] transporters were investigated. Our observations revealed that chrysin conjugates are strong inhibitors of certain biotransformation enzymes (e.g., CYP2C9) and transporters (e.g., OATP1B1, OATP1B3, OATP2B1, and BCRP) examined. Therefore, the simultaneous administration of chrysin-containing dietary supplements with medications needs to be carefully considered due to the possible development of pharmacokinetic interactions. SIGNIFICANCE STATEMENT: Chrysin-7-sulfate and chrysin-7-glucuronide are the major metabolites of flavonoid chrysin. In this study, we examined the effects of chrysin and its conjugates on cytochrome P450 enzymes and on organic anion-transporting polypeptides and ATP binding cassette transporters (P-glycoprotein, breast cancer resistance protein, and multidrug resistance-associated protein 2). Our results demonstrate that chrysin and/or its conjugates can significantly inhibit some of these proteins. Since chrysin is also contained by dietary supplements, high intake of chrysin may interrupt the transport and/or the biotransformation of drugs.
Collapse
Affiliation(s)
- Violetta Mohos
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Orsolya Ungvári
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Éva Bakos
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Katalin Kuffa
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Tímea Bencsik
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Balázs Zoltán Zsidó
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Csaba Hetényi
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Ágnes Telbisz
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy (V.M., E.F.-N., M.P.), János Szentágothai Research Centre (V.M., E.F.-N., M.P.), Department of Pharmacognosy, Faculty of Pharmacy (T.B.), and Department of Pharmacology and Pharmacotherapy, Medical School (B.Z.Z., C.H.), University of Pécs, Pécs, Hungary; and Membrane Protein Research Group (O.U., É.B., C.Ö.-L.) and Biomembrane Research Group (K.K., Á.T.), Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
30
|
Zhang Y, Holenarsipur VK, Kandoussi H, Zeng J, Mariappan TT, Sinz M, Shen H. Detection of Weak Organic Anion-Transporting Polypeptide 1B Inhibition by Probenecid with Plasma-Based Coproporphyrin in Humans. Drug Metab Dispos 2020; 48:841-848. [PMID: 32723847 DOI: 10.1124/dmd.120.000076] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/13/2020] [Indexed: 02/13/2025] Open
Abstract
Probenecid (PROB) is a clinical probe inhibitor of renal organic anion transporter (OAT) 1 and OAT3 that inhibits in vitro activity of hepatic drug transporters OATP1B1 and OATP1B3. It was hypothesized that PROB could potentially affect the disposition of OATP1B drug substrates. The plasma levels of the OATP1B endogenous biomarker candidates, including coproporphyrin I (CPI), CPIII, hexadecanedioate (HDA), and tetradecanedioate (TDA), were examined in 14 healthy subjects treated with PROB. After oral administration with 1000 mg PROB alone and in combination with furosemide (FSM), AUC (0-24 h) values were 1.39 ± 0.21-fold and 1.57 ± 0.41-fold higher than predose levels for CPI and 1.34 ± 0.16-fold and 1.45 ± 0.57-fold higher for CPIII. Despite increased systemic exposures, no decreases in CPI and CPIII renal clearance were observed (0.97 ± 0.38-fold and 1.16 ± 0.51-fold for CPI, and 1.34 ± 0.53-fold and 1.50 ± 0.69-fold for CPIII, respectively). These results suggest that the increase of CP systemic exposure is caused by OATP1B inhibition. Consistent with this hypothesis, PROB inhibited OATP1B1- and OATP1B3-mediated transport of CPI in a concentration-dependent manner, with IC50 values of 167 ± 42.0 and 76.0 ± 17.2 µM, respectively, in transporter-overexpressing human embryonic kidney cell assay. The inhibition potential was further confirmed by CPI and CPIII hepatocyte uptake experiments. In contrast, administration of PROB alone did not change AUC (0-24 h) of HDA and TDA relative to prestudy levels, although the administration of PROB in combination with FSM increased HDA and TDA levels compared with FSM alone (1.02 ± 0.18-fold and 0.90 ± 0.20-fold vs. 1.71 ± 0.43-fold and 1.62 ± 0.40-fold). Taken together, these findings indicate that PROB displays weak OATP1B inhibitory effects in vivo and that coproporphyrin is a sensitive endogenous probe of OATP1B inhibition. This study provides an explanation for the heretofore unknown mechanism responsible for PROB's interaction with other xenobiotics. SIGNIFICANCE STATEMENT: This study suggested that PROB is a weak clinical inhibitor of OATP1B based on the totality of evidence from the clinical interaction between PROB and CP and the in vitro inhibitory effect of PROB on OATP1B-mediated CP uptake. It demonstrates a new methodology of utilizing endogenous biomarkers to evaluate complex drug-drug interaction, providing explanation for the heretofore unknown mechanism responsible for PROB's inhibition. It provides evidence to strengthen the claim that CP is a sensitive circulating endogenous biomarker of OATP1B inhibition.
Collapse
Affiliation(s)
- Yueping Zhang
- Departments of Metabolism and Pharmacokinetics (Y.Z., M.S., H.S.) and Bioanalytical Sciences (H.K., J.Z.), Bristol Myers Squibb Company, Princeton, New Jersey; and Departments of Metabolism and Pharmacokinetics (V.K.H., T.T.M.), Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Biocon Park, Bommasandra IV Phase, Bangalore, India
| | - Vinay K Holenarsipur
- Departments of Metabolism and Pharmacokinetics (Y.Z., M.S., H.S.) and Bioanalytical Sciences (H.K., J.Z.), Bristol Myers Squibb Company, Princeton, New Jersey; and Departments of Metabolism and Pharmacokinetics (V.K.H., T.T.M.), Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Biocon Park, Bommasandra IV Phase, Bangalore, India
| | - Hamza Kandoussi
- Departments of Metabolism and Pharmacokinetics (Y.Z., M.S., H.S.) and Bioanalytical Sciences (H.K., J.Z.), Bristol Myers Squibb Company, Princeton, New Jersey; and Departments of Metabolism and Pharmacokinetics (V.K.H., T.T.M.), Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Biocon Park, Bommasandra IV Phase, Bangalore, India
| | - Jianing Zeng
- Departments of Metabolism and Pharmacokinetics (Y.Z., M.S., H.S.) and Bioanalytical Sciences (H.K., J.Z.), Bristol Myers Squibb Company, Princeton, New Jersey; and Departments of Metabolism and Pharmacokinetics (V.K.H., T.T.M.), Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Biocon Park, Bommasandra IV Phase, Bangalore, India
| | - T Thanga Mariappan
- Departments of Metabolism and Pharmacokinetics (Y.Z., M.S., H.S.) and Bioanalytical Sciences (H.K., J.Z.), Bristol Myers Squibb Company, Princeton, New Jersey; and Departments of Metabolism and Pharmacokinetics (V.K.H., T.T.M.), Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Biocon Park, Bommasandra IV Phase, Bangalore, India
| | - Michael Sinz
- Departments of Metabolism and Pharmacokinetics (Y.Z., M.S., H.S.) and Bioanalytical Sciences (H.K., J.Z.), Bristol Myers Squibb Company, Princeton, New Jersey; and Departments of Metabolism and Pharmacokinetics (V.K.H., T.T.M.), Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Biocon Park, Bommasandra IV Phase, Bangalore, India
| | - Hong Shen
- Departments of Metabolism and Pharmacokinetics (Y.Z., M.S., H.S.) and Bioanalytical Sciences (H.K., J.Z.), Bristol Myers Squibb Company, Princeton, New Jersey; and Departments of Metabolism and Pharmacokinetics (V.K.H., T.T.M.), Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Biocon Park, Bommasandra IV Phase, Bangalore, India
| |
Collapse
|
31
|
Prediction of Cyclosporin-Mediated Drug Interaction Using Physiologically Based Pharmacokinetic Model Characterizing Interplay of Drug Transporters and Enzymes. Int J Mol Sci 2020; 21:ijms21197023. [PMID: 32987693 PMCID: PMC7582433 DOI: 10.3390/ijms21197023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/13/2020] [Accepted: 09/18/2020] [Indexed: 12/19/2022] Open
Abstract
Uptake transporter organic anion transporting polypeptides (OATPs), efflux transporters (P-gp, BCRP and MRP2) and cytochrome P450 enzymes (CYP450s) are widely expressed in the liver, intestine or kidney. They coordinately work to control drug disposition, termed as "interplay of transporters and enzymes". Cyclosporine A (CsA) is an inhibitor of OATPs, P-gp, MRP2, BCRP and CYP3As. Drug-drug interaction (DDI) of CsA with victim drugs occurs via disordering interplay of transporters and enzymes. We aimed to establish a whole-body physiologically-based pharmacokinetic (PBPK) model which predicts disposition of CsA and nine victim drugs including atorvastatin, cerivastatin, pravastatin, rosuvastatin, fluvastatin, simvastatin, lovastatin, repaglinide and bosentan, as well as drug-drug interactions (DDIs) of CsA with nine victim drugs to investigate the integrated effect of enzymes and transporters in liver, intestinal and kidney on drug disposition. Predictions were compared with observations. Most of the predictions were within 0.5-2.0 folds of observations. Atorvastatin was represented to investigate individual contributions of transporters and CYP3As to atorvastatin disposition and their integrated effect. The contributions to atorvastatin disposition were hepatic OATPs >> hepatic CYP3A > intestinal CYP3As ≈ efflux transporters (P-gp/BCRP/MRP2). The results got the conclusion that the developed PBPK model characterizing the interplay of enzymes and transporters was successfully applied to predict the pharmacokinetics of 10 OATP substrates and DDIs of CsA with 9 victim drugs.
Collapse
|
32
|
McFeely SJ, Ritchie TK, Yu J, Nordmark A, Berglund EG, Levy RH, Ragueneau‐Majlessi I. Inhibitors of Organic Anion‐Transporting Polypeptides 1B1 and 1B3: Clinical Relevance and Regulatory Perspective. J Clin Pharmacol 2020; 60:1087-1098. [DOI: 10.1002/jcph.1604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/11/2020] [Indexed: 12/22/2022]
Affiliation(s)
| | - Tasha K. Ritchie
- University of Washington Drug Interaction Solutions Seattle Washington USA
| | - Jingjing Yu
- University of Washington Drug Interaction Solutions Seattle Washington USA
| | | | - Eva Gil Berglund
- Certara Strategic ConsultingIntegrated Drug Development Oss The Netherlands
| | - Rene H. Levy
- University of Washington Drug Interaction Solutions Seattle Washington USA
| | | |
Collapse
|
33
|
Nozaki Y, Izumi S. Recent advances in preclinical in vitro approaches towards quantitative prediction of hepatic clearance and drug-drug interactions involving organic anion transporting polypeptide (OATP) 1B transporters. Drug Metab Pharmacokinet 2020; 35:56-70. [DOI: 10.1016/j.dmpk.2019.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/29/2019] [Accepted: 11/02/2019] [Indexed: 12/26/2022]
|
34
|
Ueda K, Nakamura T, Tanaka S, Hosokawa M, Iwakawa S, Ogawara KI. Numerical analysis of apparent decitabine uptake in HCT116 cells: Incorporation of a bidirectional first-order kinetic parameter for ENT1 transport and Michaelis-Menten parameters for subsequent phosphorylation. Drug Metab Pharmacokinet 2019; 35:124-130. [PMID: 31964620 DOI: 10.1016/j.dmpk.2019.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/02/2019] [Accepted: 10/10/2019] [Indexed: 12/21/2022]
Abstract
Decitabine (DAC), a DNA methylation inhibitor, is transported into cancer cells mainly via equilibrative nucleoside transporter 1 (ENT1) and subsequently phosphorylated by deoxycytidine kinase (dCK). We previously reported that apparent DAC uptake into cells may be described using a simple compartment model with clearance for facilitated diffusion (PS) and subsequent phosphorylation (CLmet). In the present study, time course of apparent intracellular [3H]-DAC uptake was analyzed numerically, and PS and CLmet values were calculated using the compartment model in human colon cancer HCT116 cells. PS at 0.1 μM [3H]-DAC was markedly decreased in the presence of 100 μM irinotecan or etoposide, while CLmet was markedly decreased in the presence of 100 μM cytarabine or gemcitabine. CLmet at 0.1-10 μM [3H]-DAC varied in a concentration-dependent manner and was described by Michaelis-Menten parameters Km,met and Vmax,met. In conclusion, DAC uptake mainly via ENT1 may be described by a bidirectional first-order kinetic parameter, while phosphorylation by dCK may be described by Michaelis-Menten parameters.
Collapse
Affiliation(s)
- Kumiko Ueda
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| | - Touko Nakamura
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| | - Shota Tanaka
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| | - Mika Hosokawa
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| | - Seigo Iwakawa
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| | - Ken-Ichi Ogawara
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| |
Collapse
|
35
|
|
36
|
McFeely SJ, Ritchie TK, Ragueneau-Majlessi I. Variability in In Vitro OATP1B1/1B3 Inhibition Data: Impact of Incubation Conditions on Variability and Subsequent Drug Interaction Predictions. Clin Transl Sci 2019; 13:47-52. [PMID: 31468718 PMCID: PMC6951852 DOI: 10.1111/cts.12691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/08/2019] [Indexed: 12/24/2022] Open
Abstract
As the research into the organic anion transporting polypeptides (OATPs) continues to grow, it is important to ensure that the data generated are accurate and reproducible. In the in vitro evaluation of OATP1B1/1B3 inhibition, there are many variables that can contribute to variability in the resulting inhibition constants, which can then, in turn, contribute to variable results when clinical predictions (R-values) are performed. Currently, the only experimental condition recommended by the US Food and Drug Administration (FDA) is the inclusion of a pre-incubation period.1 To identify other potential sources of variability, a descriptive analysis of available in vitro inhibition data was completed. For each of the 21 substrate/inhibitor pairs evaluated, cell type and pre-incubation were found to have the greatest effect on half-maximal inhibitory concentration (IC50 ) variability. Indeed, when only HEK293 cells and co-incubation conditions were included, the observed variability for the entire data set (highest IC50 /lowest) was reduced from 12.4 to 5.2. The choice of probe substrate used in the study also had a significant effect on inhibitor constant variability. Interestingly, despite the broad range of inhibitory constants identified, these two factors showed little effect on the calculated R-values relative to the FDA evaluation cutoff of 1.1 triggering a clinical evaluation for the inhibitors evaluated. However, because of the small data set available, further research is needed to confirm these preliminary results and define best practice for the study of OATPs.
Collapse
Affiliation(s)
- Savannah J McFeely
- Department of Pharmaceutics, UW Drug Interaction Solutions, University of Washington, Seattle, Washington, USA
| | - Tasha K Ritchie
- Department of Pharmaceutics, UW Drug Interaction Solutions, University of Washington, Seattle, Washington, USA
| | - Isabelle Ragueneau-Majlessi
- Department of Pharmaceutics, UW Drug Interaction Solutions, University of Washington, Seattle, Washington, USA
| |
Collapse
|
37
|
Liang RF, Ge WJ, Song XM, Zhang JP, Cui WF, Zhang SF, Li GS. Involvement of organic anion-transporting polypeptides and organic cation transporter in the hepatic uptake of jatrorrhizine. Xenobiotica 2019; 50:479-487. [PMID: 31368836 DOI: 10.1080/00498254.2019.1651921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Jatrorrhizine possesses a wide spectrum of pharmacological activities. However, the mechanism underlying hepatic uptake of jatrorrhizine remains unclear.Rat liver slices, isolated rat hepatocytes and human embryonic kidney 293 (HEK293) cells stably expressing human organic anion-transporting polypeptide (OATP) and organic cation transporter (OCT) were used to evaluate the hepatic uptake of jatrorrhizine in this study.Uptake of jatrorrhizine in rat liver slices and isolated rat hepatocytes was significantly inhibited by glycyrrhizic acid (Oatp1b2 inhibitor) and prazosin (Oct1 inhibitor), but not by ibuprofen (Oatp1a1 inhibitor) or digoxin (Oatp1a4 inhibitor). Uptake of jatrorrhizine in OATP1B3 and OCT1-HEK293 cells indicated a saturable process with the Km of 8.20 ± 1.28 and 4.94 ± 0.55 μM, respectively. However, the transcellular transport of jatrorrhizine in OATP1B1-HEK293 cells was not observed. Rifampicin (OATP inhibitor) for OATP1B3-HEK293 cells and prazosin for OCT1-HEK293 cells could inhibit the uptake of jatrorrhizine with the IC50 of 5.49 ± 1.05 and 2.77 ± 0.72 μM, respectively.The above data indicate that hepatic uptake of jatrorrhizine is involved in both OATP and OCT, which may have important roles in jatrorrhizine liver disposition and potential drug-drug interactions.
Collapse
Affiliation(s)
- Rui-Feng Liang
- Institute of Chinese Materia Medica, Henan Provincial Academy of Traditional Chinese Medicine, Zhengzhou, China
| | - Wen-Jing Ge
- Institute of Chinese Materia Medica, Henan Provincial Academy of Traditional Chinese Medicine, Zhengzhou, China
| | - Xian-Mei Song
- Department of Immunology, Henan Medical College, Zhengzhou, China
| | - Jun-Ping Zhang
- Institute of Chinese Materia Medica, Henan Provincial Academy of Traditional Chinese Medicine, Zhengzhou, China
| | - Wei-Feng Cui
- Institute of Chinese Materia Medica, Henan Provincial Academy of Traditional Chinese Medicine, Zhengzhou, China
| | - She-Feng Zhang
- Institute of Chinese Materia Medica, Henan Provincial Academy of Traditional Chinese Medicine, Zhengzhou, China
| | - Geng-Sheng Li
- Institute of Chinese Materia Medica, Henan Provincial Academy of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
38
|
Bi YA, Costales C, Mathialagan S, West M, Eatemadpour S, Lazzaro S, Tylaska L, Scialis R, Zhang H, Umland J, Kimoto E, Tess DA, Feng B, Tremaine LM, Varma MVS, Rodrigues AD. Quantitative Contribution of Six Major Transporters to the Hepatic Uptake of Drugs: "SLC-Phenotyping" Using Primary Human Hepatocytes. J Pharmacol Exp Ther 2019; 370:72-83. [PMID: 30975793 DOI: 10.1124/jpet.119.257600] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/08/2019] [Indexed: 11/22/2022] Open
Abstract
Hepatic uptake transporters [solute carriers (SLCs)], including organic anion transporting polypeptide (OATP) 1B1, OATP1B3, OATP2B1, sodium-dependent taurocholate cotransporting polypeptide (NTCP), and organic anion (OAT2) and organic cation (OCT1) transporters, play a key role in determining the systemic and liver exposure of chemically diverse drugs. Here, we established a phenotyping approach to quantify the contribution of the six SLCs, and passive diffusion, to the overall uptake using plated human hepatocytes (PHHs). First, selective inhibitor conditions were identified by screening about 20 inhibitors across the six SLCs using single-transfected human embryonic kidney 293 cells. Data implied rifamycin SV (20 µM) inhibits three OATPs, while rifampicin (5 µM) inhibits OATP1B1/1B3 only. Further, hepatitis B virus myristoylated-preS1 peptide (0.1 µM), quinidine (100 µM), and ketoprofen (100-300 µM) are relatively selective against NTCP, OCT1, and OAT2, respectively. Second, using these inhibitory conditions, the fraction transported (ft ) by the individual SLCs was characterized for 20 substrates with PHH. Generally, extended clearance classification system class 1A/3A (e.g., warfarin) and 1B/3B compounds (e.g., statins) showed predominant OAT2 and OATP1B1/1B3 contribution, respectively. OCT1-mediated uptake was prominent for class 2/4 compounds (e.g., metformin). Third, in vitro ft values were corrected using quantitative proteomics data to obtain "scaled ft " Fourth, in vitro-in vivo extrapolation of the scaled OATP1B1/1B3 ft was assessed, leveraging statin clinical drug-drug interaction data with rifampicin as the perpetrator. Finally, we outlined a novel stepwise strategy to implement phenotypic characterization of SLC-mediated hepatic uptake for new molecular entities and drugs in a drug discovery and development setting.
Collapse
Affiliation(s)
- Yi-An Bi
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Chester Costales
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sumathy Mathialagan
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Mark West
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Soraya Eatemadpour
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sarah Lazzaro
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Laurie Tylaska
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Renato Scialis
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Hui Zhang
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - John Umland
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Emi Kimoto
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - David A Tess
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Bo Feng
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Larry M Tremaine
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Manthena V S Varma
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - A David Rodrigues
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
39
|
Amino-terminal region of human organic anion transporting polypeptide 1B1 dictates transporter stability and substrate interaction. Toxicol Appl Pharmacol 2019; 378:114642. [PMID: 31254566 DOI: 10.1016/j.taap.2019.114642] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/20/2019] [Accepted: 06/25/2019] [Indexed: 01/10/2023]
Abstract
Organic anion transporting polypeptides (OATPs) are key players of drug absorption, distribution and excretion due to their broad substrate specificity, wide tissue distribution and the involvement in drug-drug interaction. OATP1B1 is specifically localized at the basolateral membrane of human hepatocytes and serves a crucial role in the drug clearance from the body. Previous studies have shown that transmembrane domains (TMs) are essential for proper functions of OATPs. In the present study, site-directed mutagenesis was performed to study the TM1 and amino-terminus of OATP1B1. Two positively charged residues, K41 and K49, as well as a hydrophobic residue I46, in TM1 were identified to be important for the proper function of the transporter. K41A and K49A exhibited altered Km value at the high and low affinity binding sites of estrone-3- sulfate (ES), respectively; while alanine substitution of I46 showed altered Km and Vmax values for both binding components of ES. Additional replacement of K41 revealed that the positively charged property at this position is important for maintaining OATP1B1 protein level and function; while the specific side-group structure of lysine at position 49 is irreplaceable for the transporter activity. Conservative replacement of I46 with leucine also recovered the function of the transporter. In addition, studies of the amino-terminus of OATP1B1 revealed that residues ranging from 19 to 27 are essential for protein stability and substrate interaction. Therefore, the amino-terminal region, which includes TM1 and the amino-terminus of OATP1B1, is important for proper function of the membrane protein.
Collapse
|
40
|
Multiple circulating saponins from intravenous ShenMai inhibit OATP1Bs in vitro: potential joint precipitants of drug interactions. Acta Pharmacol Sin 2019; 40:833-849. [PMID: 30327544 DOI: 10.1038/s41401-018-0173-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/14/2018] [Indexed: 11/08/2022]
Abstract
ShenMai, an intravenous injection prepared from steamed Panax ginseng roots (Hongshen) and Ophiopogon japonicus roots (Maidong), is used as an add-on therapy for coronary artery disease and cancer; saponins are its bioactive constituents. Since many saponins inhibit human organic anion-transporting polypeptides (OATP)1B, this investigation determined the inhibition potencies of circulating ShenMai saponins on the transporters and the joint potential of these compounds for ShenMai-drug interaction. Circulating saponins and their pharmacokinetics were characterized in rats receiving a 30-min infusion of ShenMai at 10 mL/kg. Inhibition of human OATP1B1/1B3 and rat Oatp1b2 by the individual saponins was investigated in vitro; the compounds' joint inhibition was also assessed in vitro and the data was processed using the Chou-Talalay method. Plasma protein binding was assessed by equilibrium dialysis. Altogether, 49 saponins in ShenMai were characterized and graded into: 10-100 μmol/day (compound doses from ShenMai; 7 compounds), 1-10 μmol/day (17 compounds), and <1 μmol/day (25 compounds, including Maidong ophiopogonins). After dosing, circulating saponins were protopanaxadiol-type ginsenosides Rb1, Rb2, Rc, Rd, Ra1, Rg3, Ra2, and Ra3, protopanaxatriol-type ginsenosides Rg1, Re, Rg2, and Rf, and ginsenoside Ro. The protopanaxadiol-type ginsenosides exhibited maximum plasma concentrations of 2.1-46.6 μmol/L, plasma unbound fractions of 0.4-1.0% and terminal half-lives of 15.6-28.5 h (ginsenoside Rg3, 1.9 h), while the other ginsenosides exhibited 0.1-7.7 μmol/L, 20.8-99.2%, and 0.2-0.5 h, respectively. The protopanaxadiol-type ginsenosides, ginsenosides without any sugar attachment at C-20 (except ginsenoside Rf), and ginsenoside Ro inhibited OATP1B3 more potently (IC50, 0.2-3.5 µmol/L) than the other ginsenosides (≥22.6 µmol/L). Inhibition of OATP1B1 by ginsenosides was less potent than OATP1B3 inhibition. Ginsenosides Rb1, Rb2, Rc, Rd, Ro, Ra1, Re, and Rg2 likely contribute the major part of OATP1B3-mediated ShenMai-drug interaction potential, in an additive and time-related manner.
Collapse
|
41
|
Zhang Y, Panfen E, Fancher M, Sinz M, Marathe P, Shen H. Dissecting the Contribution of OATP1B1 to Hepatic Uptake of Statins Using the OATP1B1 Selective Inhibitor Estropipate. Mol Pharm 2019; 16:2342-2353. [DOI: 10.1021/acs.molpharmaceut.8b01226] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yueping Zhang
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Erika Panfen
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Marcus Fancher
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Michael Sinz
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Punit Marathe
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Hong Shen
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| |
Collapse
|
42
|
Malinen MM, Kauttonen A, Beaudoin JJ, Sjöstedt N, Honkakoski P, Brouwer KLR. Novel in Vitro Method Reveals Drugs That Inhibit Organic Solute Transporter Alpha/Beta (OSTα/β). Mol Pharm 2019; 16:238-246. [PMID: 30481467 PMCID: PMC6465078 DOI: 10.1021/acs.molpharmaceut.8b00966] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Drug interactions with the organic solute transporter alpha/beta (OSTα/β) are understudied even though OSTα/β is an important transporter that is expressed in multiple human tissues including the intestine, kidneys, and liver. In this study, an in vitro method to identify novel OSTα/β inhibitors was first developed using OSTα/β-overexpressing Flp-In 293 cells. Incubation conditions were optimized using previously reported OSTα/β inhibitors. A method including a 10 min preincubation step with the test compound was used to screen for OSTα/β inhibition by 77 structurally diverse compounds and fixed-dose combinations. Seven compounds and one fixed-dose combination (100 μM final concentration) inhibited OSTα/β-mediated dehydroepiandrosterone sulfate (DHEAS) uptake by >25%. Concentration-dependent OSTα/β inhibition was evaluated for all putative inhibitors (atorvastatin, ethinylestradiol, fidaxomicin, glycochenodeoxycholate, norgestimate, troglitazone, and troglitazone sulfate). Ethinylestradiol, fidaxomicin, and troglitazone sulfate yielded a clear concentration-inhibition response with IC50 values <200 μM. Among all tested compounds, there was no clear association between physicochemical properties, the severity of hepatotoxicity, and the degree of OSTα/β inhibition. This study utilized a novel in vitro method to identify OSTα/β inhibitors and, for the first time, provided IC50 values for OSTα/β inhibition. These data provide evidence that several drugs, some of which are associated with cholestatic drug-induced liver injury, may impair the function of the OSTα/β transporter.
Collapse
Affiliation(s)
- Melina M. Malinen
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Antti Kauttonen
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - James J. Beaudoin
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Noora Sjöstedt
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Paavo Honkakoski
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kim L. R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
43
|
Jójárt R, Pécsy S, Keglevich G, Szécsi M, Rigó R, Özvegy-Laczka C, Kecskeméti G, Mernyák E. Pd-Catalyzed microwave-assisted synthesis of phosphonated 13α-estrones as potential OATP2B1, 17β-HSD1 and/or STS inhibitors. Beilstein J Org Chem 2018; 14:2838-2845. [PMID: 30498534 PMCID: PMC6244214 DOI: 10.3762/bjoc.14.262] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/30/2018] [Indexed: 01/13/2023] Open
Abstract
Novel 2- or 4-phosphonated 13α-estrone derivatives were synthesized via the Hirao reaction. Bromo regioisomers (2- or 4-) of 13α-estrone and its 3-benzyl or 3-methyl ether were reacted with diethyl phosphite or diphenylphosphine oxide using Pd(PPh3)4 as catalyst under microwave irradiation. The influence of the new compounds on the transport function of the organic anion transporting polypeptide OATP2B1 was investigated by measuring Cascade Blue uptake. Derivatives bearing a 3-benzyl ether function displayed substantial submicromolar OATP2B1 inhibitory activity. The inhibitory effects of the compounds on human placental steroid sulfatase (STS) and 17β-hydroxysteroid dehydrogenase type 1 isozyme (17β-HSD1) were investigated by in vitro radiosubstrate incubation methods. None of the test compounds inhibited the STS markedly. The structure-activity relationship evaluation revealed that 2-substituted 3-hydroxy derivatives are able to inhibit the 17β-HSD1 enzyme with submicromolar IC50 values. Dual OATP2B1 and 17β-HSD1 inhibitors have been identified.
Collapse
Affiliation(s)
- Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Szabolcs Pécsy
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - György Keglevich
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1521 Budapest, Hungary
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720 Szeged, Hungary
| | - Réka Rigó
- Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Membrane protein research group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Gábor Kecskeméti
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| |
Collapse
|
44
|
Uchiyama H, Tsujimoto M, Kimura A, Yuki E, Saiki T, Yoshida T, Furukubo T, Izumi S, Yamakawa T, Tachiki H, Minegaki T, Nishiguchi K. Effects of Uremic Serum Residue on OATP1B1- and OATP1B3-Mediated Pravastatin Uptake in OATP-Expressing HEK293 Cells and Human Hepatocytes. Ther Apher Dial 2018; 23:126-132. [PMID: 30318712 DOI: 10.1111/1744-9987.12758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022]
Abstract
Patients with end-stage renal disease have increased plasma concentrations of statins, which is a risk factor for rhabdomyolysis, as well as elevated levels of uremic toxins (UTs). We investigated the effects of uremic serum residue and UTs on organic anion-transporting peptide (OATP1B1)- and OATP1B3-mediated pravastatin uptake. We evaluated the effects of normal serum residue with four UTs (hippuric acid, 3-carboxy-4-methyl-5-propyl-2-furan propionate, indole-3-acetic acid, and 3-indoxyl sulfate) and uremic serum residue on pravastatin uptake by OATP1B1- or OATP1B3-expressing HEK293 cells. Furthermore, we assessed the contribution of each transporter using cryopreserved human hepatocytes. Uremic serum residue and UTs significantly inhibited OATP1B1-mediated pravastatin uptake. Uremic serum residue accelerated OATP1B3-mediated pravastatin uptake, while UTs had no effect. There was no difference in pravastatin uptake between uremic- and normal serum residue-treated hepatocytes. The results suggest that the effects of uremic serum on pravastatin hepatic uptake may be considered negligible in end-stage renal disease patients.
Collapse
Affiliation(s)
- Hitoshi Uchiyama
- Scientific Research and Business Development Department, Towa Pharmaceutical Co. Ltd
| | - Masayuki Tsujimoto
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Akari Kimura
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Eriko Yuki
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Takashi Saiki
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Takuya Yoshida
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan.,Department of Pharmacy Service, Shirasagi Hospital, Osaka, Japan
| | - Taku Furukubo
- Department of Pharmacy Service, Shirasagi Hospital, Osaka, Japan
| | - Satoshi Izumi
- Department of Pharmacy Service, Shirasagi Hospital, Osaka, Japan
| | | | - Hidehisa Tachiki
- Scientific Research and Business Development Department, Towa Pharmaceutical Co. Ltd
| | - Tetsuya Minegaki
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kohshi Nishiguchi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
45
|
Bacsa I, Herman BE, Jójárt R, Herman KS, Wölfling J, Schneider G, Varga M, Tömböly C, Rižner TL, Szécsi M, Mernyák E. Synthesis and structure-activity relationships of 2- and/or 4-halogenated 13β- and 13α-estrone derivatives as enzyme inhibitors of estrogen biosynthesis. J Enzyme Inhib Med Chem 2018; 33:1271-1282. [PMID: 30230387 PMCID: PMC6147116 DOI: 10.1080/14756366.2018.1490731] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Ring A halogenated 13α-, 13β-, and 17-deoxy-13α-estrone derivatives were synthesised with N-halosuccinimides as electrophile triggers. Substitutions occurred at positions C-2 and/or C-4. The potential inhibitory action of the halogenated estrones on human aromatase, steroid sulfatase, or 17β-hydroxysteroid dehydrogenase 1 activity was investigated via in vitro radiosubstrate incubation. Potent submicromolar or low micromolar inhibitors were identified with occasional dual or multiple inhibitory properties. Valuable structure–activity relationships were established from the comparison of the inhibitory data obtained. Kinetic experiments performed with selected compounds revealed competitive reversible inhibition mechanisms against 17β-hydroxysteroid dehydrogenase 1 and competitive irreversible manner in the inhibition of the steroid sulfatase enzyme.
Collapse
Affiliation(s)
- Ildikó Bacsa
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | | | - Rebeka Jójárt
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | | | - János Wölfling
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | - Gyula Schneider
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | - Mónika Varga
- c Department of Microbiology , University of Szeged, University of Szeged , Szeged , Hungary
| | - Csaba Tömböly
- d Laboratory of Chemical Biology , Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences , Szeged , Hungary
| | - Tea Lanišnik Rižner
- e Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| | - Mihály Szécsi
- b 1st Department of Medicine , University of Szeged , Szeged , Hungary
| | - Erzsébet Mernyák
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| |
Collapse
|
46
|
Liao M, Zhu Q, Zhu A, Gemski C, Ma B, Guan E, Li AP, Xiao G, Xia CQ. Comparison of uptake transporter functions in hepatocytes in different species to determine the optimal model for evaluating drug transporter activities in humans. Xenobiotica 2018; 49:852-862. [PMID: 30132394 DOI: 10.1080/00498254.2018.1512017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A thorough understanding of species-dependent differences in hepatic uptake transporters is critical for predicting human pharmacokinetics (PKs) from preclinical data. In this study, the activities of organic anion transporting polypeptide (OATP/Oatp), organic cation transporter 1 (OCT1/Oct1), and sodium-taurocholate cotransporting polypeptide (NTCP/Ntcp) in cultured rat, dog, monkey and human hepatocytes were compared. The activities of hepatic uptake transporters were evaluated with respect to culture duration, substrate and species-dependent differences in hepatocytes. Longer culture duration reduced hepatic uptake transporter activities across species except for Oatp and Ntcp in rats. Comparable apparent Michaelis-Menten constant (Km,app) values in hepatocytes were observed across species for atorvastatin, estradiol-17β-glucuronide and metformin. The Km,app values for rosuvastatin and taurocholate were significantly different across species. Rat hepatocytes exhibited the highest Oatp percentage of uptake transporter-mediated permeation clearance (PSinf,act) while no difference in %PSinf,act of probe substrates were observed across species. The in vitro hepatocyte inhibition data in rats, monkeys and humans provided reasonable predictions of in vivo drug-drug interaction (DDIs) between atorvastatin/rosuvastatin and rifampin. These findings suggested that using human hepatocytes with a short culture time is the most robust preclinical model for predicting DDIs for compounds exhibiting active hepatic uptake in humans.
Collapse
Affiliation(s)
| | - Qing Zhu
- b Takeda Pharmaceuticals International Co , Cambridge , MA , USA
| | - Andy Zhu
- b Takeda Pharmaceuticals International Co , Cambridge , MA , USA
| | | | - Bingli Ma
- b Takeda Pharmaceuticals International Co , Cambridge , MA , USA
| | - Emily Guan
- a Takeda Pharmaceuticals, DMPK , Cambridge , MA , USA
| | | | - Guangqing Xiao
- b Takeda Pharmaceuticals International Co , Cambridge , MA , USA
| | - Cindy Q Xia
- b Takeda Pharmaceuticals International Co , Cambridge , MA , USA
| |
Collapse
|
47
|
Kosa RE, Lazzaro S, Bi YA, Tierney B, Gates D, Modi S, Costales C, Rodrigues AD, Tremaine LM, Varma MV. Simultaneous Assessment of Transporter-Mediated Drug-Drug Interactions Using a Probe Drug Cocktail in Cynomolgus Monkey. Drug Metab Dispos 2018; 46:1179-1189. [PMID: 29880631 DOI: 10.1124/dmd.118.081794] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
We aim to establish an in vivo preclinical model to enable simultaneous assessment of inhibition potential of an investigational drug on clinically relevant drug transporters, organic anion-transporting polypeptide (OATP)1B, breast cancer resistance protein (BCRP), P-glycoprotein (P-gp), and organic anion transporter (OAT)3. Pharmacokinetics of substrate cocktail consisting of pitavastatin (OATP1B substrate), rosuvastatin (OATP1B/BCRP/OAT3), sulfasalazine (BCRP), and talinolol (P-gp) were obtained in cynomolgus monkey-alone or in combination with transporter inhibitors. Single-dose rifampicin (30 mg/kg) significantly (P < 0.01) increased the plasma exposure of all four drugs, with a marked effect on pitavastatin and rosuvastatin [area under the plasma concentration-time curve (AUC) ratio ∼21-39]. Elacridar, BCRP/P-gp inhibitor, increased the AUC of sulfasalazine, talinolol, as well as rosuvastatin and pitavastatin. An OAT1/3 inhibitor (probenecid) significantly (P < 0.05) impacted the renal clearance of rosuvastatin (∼8-fold). In vitro, rifampicin (10 µM) inhibited uptake of pitavastatin, rosuvastatin, and sulfasalazine by monkey and human primary hepatocytes. Transport studies using membrane vesicles suggested that all probe substrates, except talinolol, are transported by cynoBCRP, whereas talinolol is a cynoP-gp substrate. Elacridar and rifampicin inhibited both cynoBCRP and cynoP-gp in vitro, indicating potential for in vivo intestinal efflux inhibition. In conclusion, a probe substrate cocktail was validated to simultaneously evaluate perpetrator impact on multiple clinically relevant transporters using the cynomolgus monkey. The results support the use of the cynomolgus monkey as a model that could enable drug-drug interaction risk assessment, before advancing a new molecular entity into clinical development, as well as providing mechanistic insights on transporter-mediated interactions.
Collapse
Affiliation(s)
- Rachel E Kosa
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Sarah Lazzaro
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Yi-An Bi
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Brendan Tierney
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Dana Gates
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Sweta Modi
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Chester Costales
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - A David Rodrigues
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Larry M Tremaine
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| | - Manthena V Varma
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design (R.E.K., S.L., Y.-a.B., B.T., C.C., A.D.R., L.M.T., M.V.V.) and Research Formulations, Pharmaceutical Sciences (D.G., S.M.), Pfizer Worldwide R&D, Groton, Connecticut
| |
Collapse
|
48
|
Dolberg AM, Reichl S. Expression analysis of human solute carrier (SLC) family transporters in nasal mucosa and RPMI 2650 cells. Eur J Pharm Sci 2018; 123:277-294. [PMID: 30041030 DOI: 10.1016/j.ejps.2018.07.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/24/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
With nearly 400 members, the solute-linked carrier (SLC) superfamily is one of the most important gene classes concerning the disposition of drugs and the transport of physiological substrates in the human body. The mapping of related transport proteins is already well advanced for the intestines, kidneys and liver, but it has recently been brought into focus for various respiratory epithelia. The aim of this study was to evaluate the expression of several SLC transporters in differently cultured RPMI 2650 cells, as well as in specimens of the human nasal mucosa. The expression profiles of PEPT2, OATP1A2, OATP4C1, OCT2, OCTN1 and OCTN2 were investigated at the gene and protein levels by performing RT-PCR, western blot analysis and immunohistological staining. Uptake assays using appropriate substrates and inhibitory substances were performed to compare the activity of peptide, organic anion and organic cation transporters, respectively, among the three models. Expression of the six SLC transporters under investigation was confirmed at the mRNA and protein levels in human nasal mucosa ex vivo as well as in RPMI 2650 cells grown under different culture conditions. The functionality was almost equal among all of the models for the PEPT and OCT(N) transporters, while the functional activity of the OATP transporters was more pronounced for both in vitro models than for excised nasal tissue. Despite negligible variations in transporter capacities, the RPMI 2650 cell cultures and freshly isolated human nasal epithelium showed nearly comparable expression patterns for the examined SLC proteins. Therefore, in vitro models based on the RPMI 2650 cell line could provide helpful data during the preclinical investigation of intranasally administered drug formulations and in the development of strategies to target nasal drug transporters for either local or systemic drug delivery.
Collapse
Affiliation(s)
- Anne M Dolberg
- Institut für Pharmazeutische Technologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Stephan Reichl
- Institut für Pharmazeutische Technologie, Technische Universität Braunschweig, Braunschweig, Germany; Zentrum für Pharmaverfahrenstechnik, Technische Universität Braunschweig, Braunschweig, Germany.
| |
Collapse
|
49
|
Yao Y, Toshimoto K, Kim SJ, Yoshikado T, Sugiyama Y. Quantitative Analysis of Complex Drug-Drug Interactions between Cerivastatin and Metabolism/Transport Inhibitors Using Physiologically Based Pharmacokinetic Modeling. Drug Metab Dispos 2018; 46:924-933. [PMID: 29712725 DOI: 10.1124/dmd.117.079210] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/25/2018] [Indexed: 02/06/2023] Open
Abstract
Cerivastatin (CER) was withdrawn from the world market because of lethal rhabdomyolysis. Coadministrations of CER and cyclosporine A (CsA) or gemfibrozil (GEM) have been reported to increase the CER blood concentration. CsA is an inhibitor of organic anion transporting polypeptide (OATP)1B1 and CYP3A4, and GEM and its glucuronide (GEM-glu) inhibit OATP1B1 and CYP2C8. The purpose of this study was to describe the transporter-/enzyme-mediated drug-drug interactions (DDIs) of CER with CsA or GEM based on unified physiologically based pharmacokinetic (PBPK) models and to investigate whether the DDIs can be quantitatively analyzed by a bottom-up approach. Initially, the PBPK models for CER and GEM/GEM-glu were constructed based on the previously reported standard protocols. Next, the drug-dependent parameters were optimized by Cluster Newton Method. Thus, described concentration-time profiles for CER and GEM/GEM-glu agreed well with the clinically observed data. The DDIs were then simulated using the established PBPK models with previously obtained in vitro inhibition constants of CsA or GEM/GEM-glu against the OATP1B1 and cytochrome P450s. DDIs with the inhibitors were underestimated compared with observed data using the geometric means of reported values. To search for better described parameters within the range of in vitro values, sensitivity analyses were performed for DDIs of CER. Using the in vitro parameter sets selected by sensitivity analyses, these DDIs were well reproduced, indicating that the present PBPK models were able to describe adequately the clinical DDIs based on a bottom-up approach. The approaches in this study would be applicable to the prediction of other DDIs involving both transporters and metabolic enzymes.
Collapse
Affiliation(s)
- Yoshiaki Yao
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Kota Toshimoto
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Soo-Jin Kim
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Takashi Yoshikado
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Yuichi Sugiyama
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| |
Collapse
|
50
|
Xiao G, Rowbottom C, Boiselle C, Gan LS. Fampridine is a Substrate and Inhibitor of Human OCT2, but not of Human MATE1, or MATE2K. Pharm Res 2018; 35:159. [DOI: 10.1007/s11095-018-2445-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/08/2018] [Indexed: 12/22/2022]
|