1
|
Rysz MA, Schäfer AM, Kinzi J, Paloumpis N, In-Albon K, Schmidlin S, Seibert I, Ricklin D, Meyer Zu Schwabedissen HE. Erlotinib-A substrate and inhibitor of OATP2B1: pharmacokinetics and CYP3A-mediated metabolism in rSlco2b1 -/- and SLCO2B1 +/+ rats. Drug Metab Dispos 2025; 53:100069. [PMID: 40239314 DOI: 10.1016/j.dmd.2025.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025] Open
Abstract
The tyrosine kinase inhibitor erlotinib is recognized as a substrate of cytochrome P450 enzymes and drug transporters. Indeed, erlotinib's extensive metabolism to the active metabolite OSI-420 (desmethyl erlotinib) mainly involves CYP3A enzymes. Additionally, erlotinib is assumed to interact with the organic anion transporting polypeptide (OATP)2B1. In this study, we aimed to investigate the role of human OATP2B1 in erlotinib's metabolism through in vitro and in vivo experiments. Using Madin-Darby canine kidney cells expressing human OATP2B1 for competitive counterflow experiments, we confirmed erlotinib as inhibitor and substrate of the transporter. Moreover, in vitro transport experiments revealed higher cellular accumulation of erlotinib at pH 5.5 than that at pH 7.4. Pharmacokinetic evaluation of orally administered erlotinib in male SLCO2B1+/+ and rSlco2b1-/- rats revealed that the human OATP2B1 does not significantly alter serum levels of erlotinib or its main metabolite OSI-420, although we observed a longer mean residence time of the metabolite in humanized rats. Although there was no difference in the OSI-420:erlotinib ratio over time in SLCO2B1+/+ and rSlco2b1-/- rats, we assessed the role of CYP3A1 and CYP3A2 in the metabolism of erlotinib. In vitro experiments showed a contribution of both enzymes to the formation of OSI-420. For CYP3A1, we found significantly higher expression in liver microsomes of male SLCO2B1+/+ rats, while the knockout genotype showed significantly higher levels of CYP3A2. However, these differences did not affect the systemic exposure of erlotinib or OSI-420 in the rats. Our findings provide further insight into the role of OATP2B1 in the disposition of orally administered erlotinib. SIGNIFICANCE STATEMENT: This study confirms that erlotinib is a substrate of the human organic anion transporting polypeptide 2B1 transporter in vitro. In vivo experiments in rat models, however, showed no significant impact of organic anion transporting polypeptide 2B1 on the systemic exposure of erlotinib or its metabolite, OSI-420. Despite variations in CYP3A enzyme expression in SLCO2B1+/+ rats, the OSI-420:erlotinib ratio remained unchanged. Although SLCO2B1+/+ rats exhibited a longer mean residence time for OSI-420, this did not significantly alter overall exposure in orally treated animals.
Collapse
Affiliation(s)
- Marta A Rysz
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Anima M Schäfer
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jonny Kinzi
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Nikolaos Paloumpis
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Katja In-Albon
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Seraina Schmidlin
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Isabell Seibert
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Daniel Ricklin
- Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | | |
Collapse
|
2
|
Bai M, Shen Q, Wu Y, Ma Z, Wang Y, Chen M, Liu D, Zhou L. Evaluation of transport mechanisms of methotrexate in human choriocarcinoma cell lines by LC-MS/MS. J Pharm Biomed Anal 2024; 247:116268. [PMID: 38823222 DOI: 10.1016/j.jpba.2024.116268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Methotrexate (MTX) is commonly prescribed as the initial treatment for gestational trophoblastic neoplasia (GTN), but MTX monotherapy may not be effective for high-risk GTN and choriocarcinoma. The cellular uptake of MTX is essential for its pharmacological activity. Thus, our study aimed to investigate the cellular pharmacokinetics and transport mechanisms of MTX in choriocarcinoma cells. For the quantification of MTX concentrations in cellular matrix, a liquid chromatography-tandem mass spectrometry method was created and confirmed initially. MTX accumulation in BeWo, JEG-3, and JAR cells was minimal. Additionally, the mRNA levels of folate receptor α (FRα) and breast cancer resistance protein (BCRP) were relatively high in the three choriocarcinoma cell lines, whereas proton-coupled folate transporter (PCFT), reduced folate carrier (RFC), and organic anion transporter (OAT) 4 were low. Furthermore, the expression of other transporters was either very low or undetectable. Notably, the application of inhibitors and small interfering RNAs (siRNAs) targeting FRα, RFC, and PCFT led to a notable decrease in the accumulation of MTX in BeWo cells. Conversely, the co-administration of multidrug resistance protein 1 (MDR1) and BCRP inhibitors increased MTX accumulation. In addition, inhibitors of OATs and organic-anion transporting polypeptides (OATPs) reduced MTX accumulation, while peptide transporter inhibitors had no effect. Results from siRNA knockdown experiments and transporter overexpression cell models indicated that MTX was not a substrate of nucleoside transporters. In conclusion, the results indicate that FRα and multiple transporters such as PCFT, RFC, OAT4, and OATPs are likely involved in the uptake of MTX, whereas MDR1 and BCRP are implicated in the efflux of MTX from choriocarcinoma cells. These results have implications for predicting transporter-mediated drug interactions and offer potential directions for further research on enhancing MTX sensitivity.
Collapse
Affiliation(s)
- Mengru Bai
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, PR China
| | - Qian Shen
- Key Laboratory for Core Technology of Generic Drug Evaluation National Medical Product Administration, Zhejiang Institute for Food and Drug Control, Hangzhou 310052, PR China
| | - Yong Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, PR China
| | - Zhiyuan Ma
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, PR China
| | - Yuqing Wang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, PR China
| | - Mingyang Chen
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Dan Liu
- Shanghai AB Sciex Analytical Instrument Trading Co., Ltd, Shanghai 200050, PR China
| | - Lin Zhou
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, PR China.
| |
Collapse
|
3
|
Ungvári O, Bakos É, Kovacsics D, Özvegy-Laczka C. The fluorescence-based competitive counterflow assay developed for organic anion transporting polypeptides 1A2, 1B1, 1B3 and 2B1 identifies pentamidine as a selective OATP1A2 substrate. FASEB J 2023; 37:e23223. [PMID: 37781971 DOI: 10.1096/fj.202300530rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/03/2023]
Abstract
Organic anion transporting polypeptides OATP1A2, OATP1B1, OATP1B3 and OATP2B1 are Na+ - and ATP-independent exchangers of large, organic compounds, encompassing structurally diverse xenobiotics, including various drugs. These OATPs influence intestinal absorption (OATP2B1), hepatic clearance (OATP1B1/3) and blood to brain penetration (OATP1A2, OATP2B1) of their drug substrates. Consequently, OATP-mediated drug or food interactions may lead to altered pharmacokinetics and toxicity. During drug development, investigation of hepatic OATP1B1 and OATP1B3 is recommended by international regulatory agencies. Most frequently, OATP-drug interactions are investigated in an indirect assay, i.e., by examining uptake inhibition of a radioactive or fluorescent probe. However, indirect assays do not distinguish between transported substrates and non-transported OATP inhibitors. To fill this hiatus, a novel assay, termed competitive counterflow (CCF) has been developed and has since been applied for several OATPs to differentiate between substrates and non-transported inhibitors. However, previous OATP CCF assays, with the exception of that for OATP1B1, used radioactive probes. In the current study, we demonstrate that sulforhodamine 101 or pyranine can be used as fluorescent probes in a CCF assay to identify transported substrates of OATP1A2, or OATPs 1B1, 1B3 and 2B1, respectively. With the help of the newly developed fluorescence-based CCF method, we identify the FDA-approved anti-protozoal drug, pentamidine as a unique substrate of OATP1A2. Furthermore, we confirm the selective, OATP1A2-mediated uptake of pentamidine in a cytotoxicity assay. Based on our results, OATP1A2 may be an important determinant of pentamidine transport through the blood-brain barrier.
Collapse
Affiliation(s)
- Orsolya Ungvári
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Éva Bakos
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| | - Daniella Kovacsics
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| |
Collapse
|
4
|
Mairinger S, Hernández-Lozano I, Filip T, Sauberer M, Löbsch M, Stanek J, Wanek T, Sake JA, Pekar T, Ehrhardt C, Langer O. Impact of P-gp and BCRP on pulmonary drug disposition assessed by PET imaging in rats. J Control Release 2022; 349:109-117. [PMID: 35798092 DOI: 10.1016/j.jconrel.2022.06.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/21/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
Abstract
P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) are two efflux transporters which are expressed in the apical (i.e. airway lumen-facing) membranes of lung epithelial cells. To assess the influence of P-gp and BCRP on the pulmonary disposition of inhaled drugs, we performed positron emission tomography (PET) imaging in rats after intratracheal aerosolization of two model P-gp/BCRP substrate radiotracers (i.e. [11C]erlotinib and [11C]tariquidar). We studied rat groups in which both transporters were active (i.e. wild-type rats), either of the two transporters was inactive (Abcb1a/b(-/-) and Abcg2(-/-) rats) or both transporters were inactive (Abcg2(-/-) rats in which pulmonary P-gp activity was inhibited by treatment with unlabeled tariquidar). PET-measured lung distribution data were compared with brain-to-plasma radioactivity concentration ratios measured in a gamma counter at the end of the PET scan. For [11C]erlotinib, lung exposure (AUClungs) was moderately but not significantly increased in Abcb1a/b(-/-) rats (1.6-fold) and Abcg2(-/-) rats (1.5-fold), and markedly (3.6-fold, p < 0.0001) increased in tariquidar-treated Abcg2(-/-) rats, compared to wild-type rats. Similarly, the brain uptake of [11C]erlotinib was substantially (4.5-fold, p < 0.0001) increased when both P-gp and BCRP activities were impaired. For [11C]tariquidar, differences in AUClungs between groups pointed into a similar direction as for [11C]erlotinib, but were less pronounced and lacked statistical significance. Our study demonstrates functional P-gp and BCRP activity in vivo in the lungs and further suggests functional redundancy between P-gp and BCRP in limiting the pulmonary uptake of a model P-gp/BCRP substrate, analogous to the blood-brain barrier. Our results suggest that pulmonary efflux transporters are important for the efficacy and safety of inhaled drugs and that their modulation may be exploited in order to improve the pharmacokinetic and pharmacodynamic performance of pulmonary delivered drugs.
Collapse
Affiliation(s)
- Severin Mairinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Thomas Filip
- Core Facility Laboratory Animal Breeding and Husbandry, Medical University of Vienna, Vienna, Austria
| | - Michael Sauberer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Mathilde Löbsch
- Core Facility Laboratory Animal Breeding and Husbandry, Medical University of Vienna, Vienna, Austria
| | - Johann Stanek
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Wanek
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Johannes A Sake
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Thomas Pekar
- Biomedical Sciences, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
5
|
Högnäsbacka A, Poot AJ, Vugts DJ, van Dongen GAMS, Windhorst AD. The Development of Positron Emission Tomography Tracers for In Vivo Targeting the Kinase Domain of the Epidermal Growth Factor Receptor. Pharmaceuticals (Basel) 2022; 15:ph15040450. [PMID: 35455447 PMCID: PMC9033078 DOI: 10.3390/ph15040450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple small molecule PET tracers have been developed for the imaging of the epidermal growth factor receptor (EGFR). These tracers target the tyrosine kinase (TK) domain of the receptor and have been used for both quantifying EGFR expression and to differentiate between EGFR mutational statuses. However, the approaches for in vivo evaluation of these tracers are diverse and have resulted in data that are hard to compare. In this review, we analyze the historical development of the in vivo evaluation approaches, starting from the first EGFR TK PET tracer [11C]PD153035 to tracers developed based on TK inhibitors used for the clinical treatment of mutated EGFR expressing non-small cell lung cancer like [11C]erlotinib and [18F]afatinib. The evaluation of each tracer has been compiled to allow for a comparison between studies and ultimately between tracers. The main challenges for each group of tracers are thereafter discussed. Finally, this review addresses the challenges that need to be overcome to be able to efficiently drive EGFR PET imaging forward.
Collapse
Affiliation(s)
- Antonia Högnäsbacka
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
- Correspondence: (A.H.); (A.D.W.)
| | - Alex J. Poot
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | - Danielle J. Vugts
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | - Guus A. M. S. van Dongen
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | - Albert D. Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
- Correspondence: (A.H.); (A.D.W.)
| |
Collapse
|
6
|
Clinical evaluation of [18F]pitavastatin for quantitative analysis of hepatobiliary transporter activity. Drug Metab Pharmacokinet 2022; 44:100449. [DOI: 10.1016/j.dmpk.2022.100449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 11/23/2022]
|
7
|
Marie S, Hernández-Lozano I, Breuil L, Truillet C, Hu S, Sparreboom A, Tournier N, Langer O. Imaging-Based Characterization of a Slco2b1(-/-) Mouse Model Using [ 11C]Erlotinib and [ 99mTc]Mebrofenin as Probe Substrates. Pharmaceutics 2021; 13:918. [PMID: 34205780 PMCID: PMC8233734 DOI: 10.3390/pharmaceutics13060918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 01/19/2023] Open
Abstract
Organic anion-transporting polypeptide 2B1 (OATP2B1) is co-localized with OATP1B1 and OATP1B3 in the basolateral hepatocyte membrane, where it is thought to contribute to the hepatic uptake of drugs. We characterized a novel Slco2b1(-/-) mouse model using positron emission tomography (PET) imaging with [11C]erlotinib (a putative OATP2B1-selective substrate) and planar scintigraphic imaging with [99mTc]mebrofenin (an OATP1B1/1B3 substrate, which is not transported by OATP2B1). Dynamic 40-min scans were performed after intravenous injection of either [11C]erlotinib or [99mTc]mebrofenin in wild-type and Slco2b1(-/-) mice. A pharmacokinetic model was used to estimate the hepatic uptake clearance (CL1) and the rate constants for transfer of radioactivity from the liver to the blood (k2) and excreted bile (k3). CL1 was significantly reduced in Slco2b1(-/-) mice for both radiotracers (p < 0.05), and k2 was significantly lower (p < 0.01) in Slco2b1(-/-) mice for [11C]erlotinib, but not for [99mTc]mebrofenin. Our data support previous evidence that OATP transporters may contribute to the hepatic uptake of [11C]erlotinib. However, the decreased hepatic uptake of the OATP1B1/1B3 substrate [99mTc]mebrofenin in Slco2b1(-/-) mice questions the utility of this mouse model to assess the relative contribution of OATP2B1 to the liver uptake of drugs which are substrates of multiple OATPs.
Collapse
Affiliation(s)
- Solène Marie
- Laboratoire d’Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, Inserm, 4 Place du Général Leclerc, 91401 Orsay, France; (S.M.); (L.B.); (C.T.)
- Département de Pharmacie Clinique, Faculté de Pharmacie, Université Paris-Saclay, 92296 Châtenay-Malabry, France
- AP-HP, Université Paris-Saclay, Hôpital Bicêtre, Pharmacie Clinique, 94270 Le Kremlin Bicêtre, France
| | - Irene Hernández-Lozano
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (I.H.-L.); (O.L.)
| | - Louise Breuil
- Laboratoire d’Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, Inserm, 4 Place du Général Leclerc, 91401 Orsay, France; (S.M.); (L.B.); (C.T.)
| | - Charles Truillet
- Laboratoire d’Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, Inserm, 4 Place du Général Leclerc, 91401 Orsay, France; (S.M.); (L.B.); (C.T.)
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (S.H.); (A.S.)
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (S.H.); (A.S.)
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, Inserm, 4 Place du Général Leclerc, 91401 Orsay, France; (S.M.); (L.B.); (C.T.)
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (I.H.-L.); (O.L.)
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
8
|
Kinzi J, Grube M, Meyer Zu Schwabedissen HE. OATP2B1 - The underrated member of the organic anion transporting polypeptide family of drug transporters? Biochem Pharmacol 2021; 188:114534. [PMID: 33794186 DOI: 10.1016/j.bcp.2021.114534] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022]
Abstract
The organic anion transporting polypeptide 2B1 (OATP2B1) was one of the first cloned members of the SLCO family. However, its physiological and pharmacological role is still poorly understood, and object of a current debate on the transporter's relevance. Within this commentary, we summarize the data currently available on the transporter's expression and its substrates and highlight the strength and difficulties of the methods that have been applied to gather these data. The conclusion drawn from these findings was that OATP2B1 due to its intestinal expression is most likely involved in oral drug absorption of its substrate and therefore prone for interactions. This has been tested in in vivo drug interaction and/or pharmacogenetic studies. While some of these support the notion of OATP2B1 being of relevance in drug absorption, the pharmacogenetic findings are rather inconclusive. We will explain our thoughts why OATP2B1 may not influence the general systemic pharmacokinetic of certain substrates, but possibly local distribution processes, like the transfer across the blood-brain-barrier. Besides the pharmacokinetic aspects, there are data on endogenous molecules like coproporphyrins and sulfated steroids. Therefore, we will also highlight possible physiological roles of OATP2B1, which are driven by its expression pattern in the tubular cells of the kidney as well as its expression in the blood brain barrier. Finally we also deal with the advantages and disadvantages in the use of animal models to decipher the role of OATP2B1 in pharmacokinetics of its substrates and beyond.
Collapse
Affiliation(s)
- Jonny Kinzi
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Markus Grube
- Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany
| | | |
Collapse
|
9
|
Shimada T, Okano M, Yamada M, Ogawa Y, Ueda A, Nagase K, Sai Y. Administration of erlotinib in apple juice overcomes decreased absorption in a rat model of gastric acid suppression. Drug Metab Pharmacokinet 2020; 35:534-538. [PMID: 33028492 DOI: 10.1016/j.dmpk.2020.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/03/2020] [Accepted: 08/16/2020] [Indexed: 10/23/2022]
Abstract
Erlotinib shows pH-dependent solubility and its absorption is decreased in patients receiving gastric acid suppression therapy. Here, we examined whether administration of erlotinib in acidic solutions would improve its solubility and absorption characteristics. In vitro, the solubility of erlotinib in HCl solution increased with decreasing pH, and was far higher than that in tap water. The solubility in apple juice (pH 3.7) was higher than that in HCl solution of the same pH. In vivo, the absorption of erlotinib administered in tap water was decreased in omeprazole-treated (OP) rats, used as a model of gastric acid suppression, compared to control rats. In the OP rats, the plasma concentrations in the groups given erlotinib in apple juice and in HCl (pH 3.7) were significantly higher than in the tap water group in the initial phase of absorption. AUC in OP rats given erlotinib in apple juice was 1.69-fold larger than that of control rats given erlotinib in tap water, and 2.49-fold larger than that of OP rats given erlotinib in tap water. Thus, administration of erlotinib in an acidic beverage to patients receiving gastric acid suppression therapy might be effective to increase solubility and absorption.
Collapse
Affiliation(s)
- Tsutomu Shimada
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan; Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| | - Mai Okano
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| | - Momoko Yamada
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| | - Yuki Ogawa
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| | - Arimi Ueda
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan; Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| | - Katsuhiko Nagase
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan; Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| | - Yoshimichi Sai
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan; Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| |
Collapse
|
10
|
Uptake Transporters of the SLC21, SLC22A, and SLC15A Families in Anticancer Therapy-Modulators of Cellular Entry or Pharmacokinetics? Cancers (Basel) 2020; 12:cancers12082263. [PMID: 32806706 PMCID: PMC7464370 DOI: 10.3390/cancers12082263] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Solute carrier transporters comprise a large family of uptake transporters involved in the transmembrane transport of a wide array of endogenous substrates such as hormones, nutrients, and metabolites as well as of clinically important drugs. Several cancer therapeutics, ranging from chemotherapeutics such as topoisomerase inhibitors, DNA-intercalating drugs, and microtubule binders to targeted therapeutics such as tyrosine kinase inhibitors are substrates of solute carrier (SLC) transporters. Given that SLC transporters are expressed both in organs pivotal to drug absorption, distribution, metabolism, and elimination and in tumors, these transporters constitute determinants of cellular drug accumulation influencing intracellular drug concentration required for efficacy of the cancer treatment in tumor cells. In this review, we explore the current understanding of members of three SLC families, namely SLC21 (organic anion transporting polypeptides, OATPs), SLC22A (organic cation transporters, OCTs; organic cation/carnitine transporters, OCTNs; and organic anion transporters OATs), and SLC15A (peptide transporters, PEPTs) in the etiology of cancer, in transport of chemotherapeutic drugs, and their influence on efficacy or toxicity of pharmacotherapy. We further explore the idea to exploit the function of SLC transporters to enhance cancer cell accumulation of chemotherapeutics, which would be expected to reduce toxic side effects in healthy tissue and to improve efficacy.
Collapse
|
11
|
Chen M, Hu S, Li Y, Gibson AA, Fu Q, Baker SD, Sparreboom A. Role of Oatp2b1 in Drug Absorption and Drug-Drug Interactions. Drug Metab Dispos 2020; 48:419-425. [PMID: 32114507 PMCID: PMC7180048 DOI: 10.1124/dmd.119.090316] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/18/2020] [Indexed: 12/18/2022] Open
Abstract
The organic anion transporting polypeptide (OATP)2B1 is localized on the basolateral membrane of hepatocytes and is expressed in enterocytes. Based on its distribution pattern and functional similarity to OATP1B-type transporters, OATP2B1 might have a role in the absorption and disposition of a range of xenobiotics. Although several prescription drugs, including hydroxymethylglutaryl-coenzyme A-CoA reductase inhibitors (statins) such as fluvastatin, are OATP2B1 substrates in vitro, evidence supporting the in vivo relevance of this transporter remains limited, and most has relied on substrate-inhibitor interactions resulting in altered pharmacokinetic properties of the victim drugs. To address this knowledge deficit, we developed and characterized an Oatp2b1-deficient mouse model and evaluated the impact of this transporter on the absorption and disposition of fluvastatin. Consistent with the intestinal localization of Oatp2b1, we found that the genetic deletion or pharmacological inhibition of Oatp2b1 was associated with decreased absorption of fluvastatin by 2- to 3-fold. The availability of a viable Oatp2b1-deficient mouse model provides an opportunity to unequivocally determine the contribution of this transporter to the absorption and drug-drug interaction potential of drugs. SIGNIFICANCE STATEMENT: The current investigation suggests that mice deficient in Oatp2b1 provide a valuable tool to study the in vivo importance of this transporter. In addition, our studies have identified novel potent inhibitors of OATP2B1 among the class of tyrosine kinase inhibitors, a rapidly expanding class of drugs used in various therapeutic areas that may cause drug-drug interactions with OATP2B1 substrates.
Collapse
Affiliation(s)
- Mingqing Chen
- Experimental Cancer Pharmacology Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shuiying Hu
- Experimental Cancer Pharmacology Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yang Li
- Experimental Cancer Pharmacology Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Alice A Gibson
- Experimental Cancer Pharmacology Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Qiang Fu
- Experimental Cancer Pharmacology Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Sharyn D Baker
- Experimental Cancer Pharmacology Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Alex Sparreboom
- Experimental Cancer Pharmacology Laboratory, Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| |
Collapse
|
12
|
Hernández Lozano I, Langer O. Use of imaging to assess the activity of hepatic transporters. Expert Opin Drug Metab Toxicol 2020; 16:149-164. [PMID: 31951754 PMCID: PMC7055509 DOI: 10.1080/17425255.2020.1718107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Introduction: Membrane transporters of the SLC and ABC families are abundantly expressed in the liver, where they control the transfer of drugs/drug metabolites across the sinusoidal and canalicular hepatocyte membranes and play a pivotal role in hepatic drug clearance. Noninvasive imaging methods, such as PET, SPECT or MRI, allow for measuring the activity of hepatic transporters in vivo, provided that suitable transporter imaging probes are available.Areas covered: We give an overview of the working principles of imaging-based assessment of hepatic transporter activity. We discuss different currently available PET/SPECT radiotracers and MRI contrast agents and their applications to measure hepatic transporter activity in health and disease. We cover mathematical modeling approaches to obtain quantitative parameters of transporter activity and provide a critical assessment of methodological limitations and challenges associated with this approach.Expert opinion: PET in combination with pharmacokinetic modeling can be potentially applied in drug development to study the distribution of new drug candidates to the liver and their clearance mechanisms. This approach bears potential to mechanistically assess transporter-mediated drug-drug interactions, to assess the influence of disease on hepatic drug disposition and to validate and refine currently available in vitro-in vivo extrapolation methods to predict hepatic clearance of drugs.
Collapse
Affiliation(s)
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| |
Collapse
|
13
|
Recent progress in in vivo phenotyping technologies for better prediction of transporter-mediated drug-drug interactions. Drug Metab Pharmacokinet 2020; 35:76-88. [PMID: 31948854 DOI: 10.1016/j.dmpk.2019.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 12/20/2022]
Abstract
Clinical reports on transporter-mediated drug-drug interactions (TP-DDIs) have rapidly accumulated and regulatory guidance/guidelines recommend that sponsors consider performing quantitative prediction of TP-DDI risks in the process of drug development. In vitro experiments for characterizing the function of drug transporters have been established and various parameters such as the inhibition constant (Ki) of drugs and the intrinsic uptake/efflux clearance for a certain transporter can be obtained. However, many reports have indicated large discrepancies between the parameters estimated from in vitro experiments and those rationally explaining drug pharmacokinetics. Thus, it is essential to evaluate directly the function of each transporter isoform in vivo in humans. At present, several transporter substrate drugs and endogenous compounds have been recognized as probe substrates for a specific transporter and transporter function was evaluated by monitoring the plasma and urine concentration of those probes; however, few compounds specifically transported via a single transporter isoform have been found. For monitoring the intraorgan concentration of drugs, positron emission tomography can be a powerful tool and clinical examples for quantification of in vivo transporter function have been published. In this review, novel methodologies for in vivo phenotyping of transporter function are summarized.
Collapse
|
14
|
Hernández Lozano I, Bauer M, Wulkersdorfer B, Traxl A, Philippe C, Weber M, Häusler S, Stieger B, Jäger W, Mairinger S, Wanek T, Hacker M, Zeitlinger M, Langer O. Measurement of Hepatic ABCB1 and ABCG2 Transport Activity with [ 11C]Tariquidar and PET in Humans and Mice. Mol Pharm 2019; 17:316-326. [PMID: 31790256 DOI: 10.1021/acs.molpharmaceut.9b01060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
P-Glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) in the canalicular membrane of hepatocytes mediate the biliary excretion of drugs and drug metabolites. To measure hepatic ABCB1 and ABCG2 activity, we performed positron emission tomography (PET) scans with the ABCB1/ABCG2 substrate [11C]tariquidar in healthy volunteers and wild-type, Abcb1a/b(-/-), Abcg2(-/-), and Abcb1a/b(-/-)Abcg2(-/-) mice without and with coadministration of unlabeled tariquidar. PET data were analyzed with a three-compartment pharmacokinetic model. [11C]Tariquidar underwent hepatobiliary excretion in both humans and mice, and tariquidar coadministration caused a significant reduction in the rate constant for the transfer of radioactivity from the liver into bile (by -74% in humans and by -62% in wild-type mice), suggesting inhibition of canalicular efflux transporter activity. Radio-thin-layer chromatography analysis revealed that the majority of radioactivity (>87%) in the mouse liver and bile was composed of unmetabolized [11C]tariquidar. PET data in transporter knockout mice revealed that both ABCB1 and ABCG2 mediated biliary excretion of [11C]tariquidar. In vitro experiments indicated that tariquidar is not a substrate of major hepatic basolateral uptake transporters (SLCO1B1, SLCO1B3, SLCO2B1, SLC22A1, and SLC22A3). Our data suggest that [11C]tariquidar can be used to measure hepatic canalicular ABCB1/ABCG2 transport activity without a confounding effect of uptake transporters.
Collapse
Affiliation(s)
- Irene Hernández Lozano
- Department of Clinical Pharmacology , Medical University of Vienna , Vienna 1090 , Austria
| | - Martin Bauer
- Department of Clinical Pharmacology , Medical University of Vienna , Vienna 1090 , Austria
| | - Beatrix Wulkersdorfer
- Department of Clinical Pharmacology , Medical University of Vienna , Vienna 1090 , Austria
| | - Alexander Traxl
- Preclinical Molecular Imaging , AIT Austrian Institute of Technology GmbH , Seibersdorf 2444 , Austria
| | - Cécile Philippe
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy , Medical University of Vienna , Vienna 1090 , Austria
| | - Maria Weber
- Department of Clinical Pharmacology , Medical University of Vienna , Vienna 1090 , Austria
| | - Stephanie Häusler
- Department of Clinical Pharmacology and Toxicology , University Hospital Zurich, University of Zurich , Zurich 8006 , Switzerland
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology , University Hospital Zurich, University of Zurich , Zurich 8006 , Switzerland
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics , University of Vienna , Vienna 1090 , Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging , AIT Austrian Institute of Technology GmbH , Seibersdorf 2444 , Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging , AIT Austrian Institute of Technology GmbH , Seibersdorf 2444 , Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy , Medical University of Vienna , Vienna 1090 , Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology , Medical University of Vienna , Vienna 1090 , Austria
| | - Oliver Langer
- Department of Clinical Pharmacology , Medical University of Vienna , Vienna 1090 , Austria.,Preclinical Molecular Imaging , AIT Austrian Institute of Technology GmbH , Seibersdorf 2444 , Austria.,Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy , Medical University of Vienna , Vienna 1090 , Austria
| |
Collapse
|
15
|
Hernández Lozano I, Karch R, Bauer M, Blaickner M, Matsuda A, Wulkersdorfer B, Hacker M, Zeitlinger M, Langer O. Towards Improved Pharmacokinetic Models for the Analysis of Transporter-Mediated Hepatic Disposition of Drug Molecules with Positron Emission Tomography. AAPS J 2019; 21:61. [PMID: 31037511 PMCID: PMC6488550 DOI: 10.1208/s12248-019-0323-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/19/2019] [Indexed: 12/19/2022] Open
Abstract
Positron emission tomography (PET) imaging with radiolabeled drugs holds great promise to assess the influence of membrane transporters on hepatobiliary clearance of drugs. To exploit the full potential of PET, quantitative pharmacokinetic models are required. In this study, we evaluated the suitability of different compartment models to describe the hepatic disposition of [11C]erlotinib as a small-molecule model drug which undergoes transporter-mediated hepatobiliary excretion. We analyzed two different, previously published data sets in healthy volunteers, in which a baseline [11C]erlotinib PET scan was followed by a second PET scan either after oral intake of unlabeled erlotinib (300 mg) or after intravenous infusion of the prototypical organic anion-transporting polypeptide inhibitor rifampicin (600 mg). We assessed a three-compartment (3C) and a four-compartment (4C) model, in which either a sampled arterial blood input function or a mathematically derived dual input function (DIF), which takes the contribution of the portal vein to the liver blood supply into account, was used. Both models provided acceptable fits of the observed PET data in the liver and extrahepatic bile duct and gall bladder. Changes in model outcome parameters between scans were consistent with the involvement of basolateral hepatocyte uptake and canalicular efflux transporters in the hepatobiliary clearance of [11C]erlotinib. Our results demonstrated that inclusion of a DIF did not lead to substantial improvements in model fits. The models developed in this work represent a step forward in applying PET as a tool to assess the impact of hepatic transporters on drug disposition and their involvement in drug-drug interactions.
Collapse
Affiliation(s)
- Irene Hernández Lozano
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Rudolf Karch
- Centre for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Matthias Blaickner
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Akihiro Matsuda
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Beatrix Wulkersdorfer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria.
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
16
|
Bonnaventure P, Cusin F, Pastor CM. Hepatocyte Concentrations of Imaging Compounds Associated with Transporter Inhibition: Evidence in Perfused Rat Livers. Drug Metab Dispos 2019; 47:412-418. [PMID: 30674615 DOI: 10.1124/dmd.118.084624] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/18/2019] [Indexed: 12/15/2022] Open
Abstract
In the liver, several approaches are used to investigate and predict the complex issue of drug-induced transporter inhibition. These approaches include in vitro assays and pharmacokinetic models that predict how inhibitors modify the systemic and liver concentrations of the victim drugs. Imaging is another approach that shows how inhibitors might alter liver concentrations stronger than systemic concentrations. In perfused rat livers associated with a gamma counter that measures liver concentrations continuously, we previously showed how fluxes across transporters generate the hepatocyte concentrations of two clinical imaging compounds, one with a low extraction ratio [gadobenate dimeglumine (BOPTA)] and one with a high extraction ratio [mebrofenin (MEB)]. BOPTA and MEB are transported by rat organic anion transporting polypeptide and multiple resistance-associated protein 2, which are both inhibited by rifampicin. The aim of the study is to measure how rifampicin modifies the hepatocyte concentrations and membrane clearances of BOPTA and MEB and to determine whether these compounds might be used to investigate transporter-mediated drug-drug interactions in clinical studies. We show that rifampicin coperfusion greatly decreases BOPTA hepatocyte concentrations, but increases those of MEB. Rifampicin strongly decreases BOPTA hepatic clearance. In contrast, rifampicin decreases moderately MEB hepatic clearance and blocks the biliary intrinsic clearance, increasing MEB hepatocyte concentrations. In conclusion, low concentrations prevent the quantification of BOPTA biliary intrinsic clearance, while MEB is a promising imaging probe substrate to evidence transporter-mediated drug-drug interactions when inhibitors act on influx and efflux transporters.
Collapse
Affiliation(s)
- Pierre Bonnaventure
- Department of Radiology, Hôpitaux Universitaires de Genève, Geneva, Switzerland (P.B., F.C., C.M.P.); and Laboratory of Imaging Biomarkers, Centre of Research on Inflammation, Unité Mixte de Recherche 1149, Institut National de la santé et de la Recherche Médicale and University Paris Diderot, Paris, France (C.M.P.)
| | - Fabien Cusin
- Department of Radiology, Hôpitaux Universitaires de Genève, Geneva, Switzerland (P.B., F.C., C.M.P.); and Laboratory of Imaging Biomarkers, Centre of Research on Inflammation, Unité Mixte de Recherche 1149, Institut National de la santé et de la Recherche Médicale and University Paris Diderot, Paris, France (C.M.P.)
| | - Catherine M Pastor
- Department of Radiology, Hôpitaux Universitaires de Genève, Geneva, Switzerland (P.B., F.C., C.M.P.); and Laboratory of Imaging Biomarkers, Centre of Research on Inflammation, Unité Mixte de Recherche 1149, Institut National de la santé et de la Recherche Médicale and University Paris Diderot, Paris, France (C.M.P.)
| |
Collapse
|
17
|
Traxl A, Mairinger S, Filip T, Sauberer M, Stanek J, Poschner S, Jäger W, Zoufal V, Novarino G, Tournier N, Bauer M, Wanek T, Langer O. Inhibition of ABCB1 and ABCG2 at the Mouse Blood-Brain Barrier with Marketed Drugs To Improve Brain Delivery of the Model ABCB1/ABCG2 Substrate [ 11C]erlotinib. Mol Pharm 2019; 16:1282-1293. [PMID: 30694684 DOI: 10.1021/acs.molpharmaceut.8b01217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
P-Glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) are two efflux transporters at the blood-brain barrier (BBB), which effectively restrict brain distribution of diverse drugs, such as tyrosine kinase inhibitors. There is a crucial need for pharmacological ABCB1 and ABCG2 inhibition protocols for a more effective treatment of brain diseases. In the present study, seven marketed drugs (osimertinib, erlotinib, nilotinib, imatinib, lapatinib, pazopanib, and cyclosporine A) and one nonmarketed drug (tariquidar), with known in vitro ABCB1/ABCG2 inhibitory properties, were screened for their inhibitory potency at the BBB in vivo. Positron emission tomography (PET) using the model ABCB1/ABCG2 substrate [11C]erlotinib was performed in mice. Tested inhibitors were administered as i.v. bolus injections at 30 min before the start of the PET scan, followed by a continuous i.v. infusion for the duration of the PET scan. Five of the tested drugs increased total distribution volume of [11C]erlotinib in the brain ( VT,brain) compared to vehicle-treated animals (tariquidar, + 69%; erlotinib, + 19% and +23% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 22%; lapatinib, + 25%; and cyclosporine A, + 49%). For all drugs, increases in [11C]erlotinib brain distribution were lower than in Abcb1a/b(-/-)Abcg2(-/-) mice (+149%), which suggested that only partial ABCB1/ABCG2 inhibition was reached at the mouse BBB. The plasma concentrations of the tested drugs at the time of the PET scan were higher than clinically achievable plasma concentrations. Some of the tested drugs led to significant increases in blood radioactivity concentrations measured at the end of the PET scan (erlotinib, + 103% and +113% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 125%; and cyclosporine A, + 101%), which was most likely caused by decreased hepatobiliary excretion of radioactivity. Taken together, our data suggest that some marketed tyrosine kinase inhibitors may be repurposed to inhibit ABCB1 and ABCG2 at the BBB. From a clinical perspective, moderate increases in brain delivery despite the administration of high i.v. doses as well as peripheral drug-drug interactions due to transporter inhibition in clearance organs question the translatability of this concept.
Collapse
Affiliation(s)
- Alexander Traxl
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Severin Mairinger
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Thomas Filip
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Michael Sauberer
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Johann Stanek
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Stefan Poschner
- Department of Clinical Pharmacy and Diagnostics , University of Vienna , 1090 Vienna , Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics , University of Vienna , 1090 Vienna , Austria
| | - Viktoria Zoufal
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria , 3400 Klosterneuburg , Austria
| | - Nicolas Tournier
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot , CEA, Inserm, Univ. Paris Sud, CNRS, Université Paris-Saclay , 91450 Orsay , France
| | - Martin Bauer
- Department of Clinical Pharmacology , Medical University of Vienna , 1090 Vienna , Austria
| | - Thomas Wanek
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Oliver Langer
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria.,Department of Clinical Pharmacology , Medical University of Vienna , 1090 Vienna , Austria.,Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine , Medical University of Vienna , 1090 Vienna , Austria
| |
Collapse
|