1
|
Jackwerth M, Mairinger S, Zeitlinger M, Langer O. Functional MRP1 activity in human skeletal muscle. Br J Pharmacol 2025. [PMID: 40274290 DOI: 10.1111/bph.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Affiliation(s)
- Matthias Jackwerth
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - 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
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - 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
|
2
|
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
|
3
|
Hernández-Lozano I, Leterrier S, Mairinger S, Stanek J, Zacher AS, Breyer L, Hacker M, Zeitlinger M, Pahnke J, Tournier N, Wanek T, Langer O. Performance and Sensitivity of [ 99mTc]Tc-sestamibi Compared with Positron Emission Tomography Radiotracers to Measure P-glycoprotein Function in the Kidneys and Liver. Mol Pharm 2024; 21:932-943. [PMID: 38225758 PMCID: PMC10848257 DOI: 10.1021/acs.molpharmaceut.3c01036] [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: 11/02/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/17/2024]
Abstract
P-glycoprotein (P-gp, encoded in humans by the ABCB1 gene and in rodents by the Abcb1a/b genes) is a membrane transporter that can restrict the intestinal absorption and tissue distribution of many drugs and may also contribute to renal and hepatobiliary drug excretion. The aim of this study was to compare the performance and sensitivity of currently available radiolabeled P-gp substrates for positron emission tomography (PET) with the single-photon emission computed tomography (SPECT) radiotracer [99mTc]Tc-sestamibi for measuring the P-gp function in the kidneys and liver. Wild-type, heterozygous (Abcb1a/b(+/-)), and homozygous (Abcb1a/b(-/-)) Abcb1a/b knockout mice were used as models of different P-gp abundance in excretory organs. Animals underwent either dynamic PET scans after intravenous injection of [11C]N-desmethyl-loperamide, (R)-[11C]verapamil, or [11C]metoclopramide or consecutive static SPECT scans after intravenous injection of [99mTc]Tc-sestamibi. P-gp in the kidneys and liver of the mouse models was analyzed with immunofluorescence labeling and Western blotting. In the kidneys, Abcb1a/b() mice had intermediate P-gp abundance compared with wild-type and Abcb1a/b(-/-) mice. Among the four tested radiotracers, renal clearance of radioactivity (CLurine,kidney) was significantly reduced (-83%) in Abcb1a/b(-/-) mice only for [99mTc]Tc-sestamibi. Biliary clearance of radioactivity (CLbile,liver) was significantly reduced in Abcb1a/b(-/-) mice for [11C]N-desmethyl-loperamide (-47%), [11C]metoclopramide (-25%), and [99mTc]Tc-sestamibi (-79%). However, in Abcb1a/b(+/-) mice, CLbile,liver was significantly reduced (-47%) only for [99mTc]Tc-sestamibi. Among the tested radiotracers, [99mTc]Tc-sestamibi performed best in measuring the P-gp function in the kidneys and liver. Owing to its widespread clinical availability, [99mTc]Tc-sestamibi represents a promising probe substrate to assess systemic P-gp-mediated drug-drug interactions and to measure renal and hepatic P-gp function under different (patho-)physiological conditions.
Collapse
Affiliation(s)
| | - Sarah Leterrier
- Laboratoire
d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm,
Service Hospitalier Frédéric Joliot, 91401 Orsay, France
| | - Severin Mairinger
- Department
of Clinical Pharmacology, Medical University
of Vienna, 1090 Vienna, Austria
- Department
of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Johann Stanek
- Department
of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna S. Zacher
- Department
of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Lara Breyer
- Department
of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Marcus Hacker
- Department
of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Markus Zeitlinger
- Department
of Clinical Pharmacology, Medical University
of Vienna, 1090 Vienna, Austria
| | - Jens Pahnke
- Department
of Pathology, Section of Neuropathology, Translational Neurodegeneration
Research and Neuropathology Lab, University
of Oslo (UiO) and Oslo University Hospital (OUS), 0372 Oslo, Norway
- Lübeck
Institute of Experimental Dermatology (LIED), Pahnke Lab, University of Lübeck and University Medical
Center Schleswig-Holstein, 23538 Lübeck, Germany
- Department
of Pharmacology, Faculty of Medicine, University
of Latvia, 1004 Ri̅ga, Latvia
- Department
of Neurobiology, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Nicolas Tournier
- Laboratoire
d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm,
Service Hospitalier Frédéric Joliot, 91401 Orsay, France
| | - Thomas Wanek
- Department
of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Langer
- Department
of Clinical Pharmacology, Medical University
of Vienna, 1090 Vienna, Austria
- Department
of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
4
|
Breuil L, El Biali M, Vodovar D, Marie S, Auvity S, Bauer M, Goutal S, Rodrigo S, Langer O, Tournier N. Parametric Imaging of P-Glycoprotein Function at the Blood-Brain Barrier Using k E,brain-maps Generated from [ 11C]Metoclopramide PET Data in Rats, Nonhuman Primates and Humans. Mol Imaging Biol 2023; 25:1135-1141. [PMID: 37801196 DOI: 10.1007/s11307-023-01864-z] [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: 05/12/2023] [Revised: 09/01/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
PURPOSE PET imaging using [11C]metoclopramide revealed the importance of P-glycoprotein (P-gp, ABCB1) in mediating the brain-to-blood efflux of substrates across the blood-brain barrier (BBB). In this work, the elimination rate constant from the brain (kE,brain), calculated from dynamic PET images without the need for arterial blood sampling, was evaluated as an outcome parameter for the interpretation of [11C]metoclopramide PET data. PROCEDURES kE,brain parameter was obtained by linear regression of log-transformed brain time-activity curves (TACs). kE,brain values (h-1) obtained under baseline conditions were compared with values obtained after complete P-gp inhibition using tariquidar in rats (n = 4) and baboons (n = 4) or after partial inhibition using cyclosporine A in humans (n = 10). In baboons, the sensitivity of kE,brain to measure complete P-gp inhibition was compared with outcome parameters derived from kinetic modeling using a 1-tissue compartment model (1-TCM). Finally, kE,brain-maps were generated in each species using PMOD software. RESULTS The linear part of the log-transformed brain TACs occurred from 10 to 30 min after radiotracer injection in rats, from 15 to 60 min in baboons, and from 20 to 60 min in humans. P-gp inhibition significantly decreased kE,brain values by 39 ± 12% in rats (p < 0.01), by 32 ± 6% in baboons (p < 0.001), and by 37 ± 22% in humans (p < 0.001). In baboons, P-gp inhibition consistently decreased the brain-to-plasma efflux rate constant k2 (36 ± 9%, p < 0.01) leading to an increase in the total brain volume of distribution (VT, 101 ± 12%, p < 0.001). In all studied species, brain kE,brain-maps displayed decreased P-gp-mediated efflux across the BBB. CONCLUSIONS kE,brain of [11C]metoclopramide provides a simple outcome parameter to describe P-gp function in the living brain when arterial input function data are unavailable, although less sensitive than VT. kE,brain-maps represent easy to compute parametric images reflecting the effect of P-gp on [11C]metoclopramide elimination from the brain.
Collapse
Affiliation(s)
- Louise Breuil
- Inserm, CNRS, CEA, BioMaps, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Université Paris-Saclay, CEA/SHFJ, 4 Place du Général Leclerc 91400, Orsay, France
- Inserm UMR-S1144, University of Paris Cité, 75006, Paris, France
| | - Myriam El Biali
- Department of Clinical Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Dominique Vodovar
- Inserm, CNRS, CEA, BioMaps, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Université Paris-Saclay, CEA/SHFJ, 4 Place du Général Leclerc 91400, Orsay, France
- Inserm UMR-S1144, University of Paris Cité, 75006, Paris, France
| | - Solène Marie
- Inserm, CNRS, CEA, BioMaps, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Université Paris-Saclay, CEA/SHFJ, 4 Place du Général Leclerc 91400, Orsay, France
| | - Sylvain Auvity
- Inserm, CNRS, CEA, BioMaps, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Université Paris-Saclay, CEA/SHFJ, 4 Place du Général Leclerc 91400, Orsay, France
- Inserm UMR-S1144, University of Paris Cité, 75006, Paris, France
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Sébastien Goutal
- Inserm, CNRS, CEA, BioMaps, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Université Paris-Saclay, CEA/SHFJ, 4 Place du Général Leclerc 91400, Orsay, France
| | - Sebastian Rodrigo
- Inserm, CNRS, CEA, BioMaps, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Université Paris-Saclay, CEA/SHFJ, 4 Place du Général Leclerc 91400, Orsay, France
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090, Vienna, Austria
| | - Nicolas Tournier
- Inserm, CNRS, CEA, BioMaps, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Université Paris-Saclay, CEA/SHFJ, 4 Place du Général Leclerc 91400, Orsay, France.
| |
Collapse
|
5
|
Chatterjee S, Deshpande AA, Shen H. Recent advances in the in vitro and in vivo methods to assess impact of P-glycoprotein and breast cancer resistance protein transporters in central nervous system drug disposition. Biopharm Drug Dispos 2023; 44:7-25. [PMID: 36692150 DOI: 10.1002/bdd.2345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 01/25/2023]
Abstract
One challenge in central nervous system (CNS) drug discovery has been ensuring the blood-brain barrier (BBB) penetration of compounds at an efficacious concentration that provides suitable safety margins for clinical investigation. Research providing for the accurate prediction of brain penetration of compounds during preclinical discovery is important to a CNS program. In the BBB, P-glycoprotein (P-gp) (ABCB1) and breast cancer resistance protein (BCRP) (ABCG2) transporters have been demonstrated to play a major role in the active efflux of endogenous compounds and xenobiotics out of the brain microvessel cells and back to the systemic circulation. In the past 10 years, there has been significant technological improvement in the sensitivity of quantitative proteomics methods, in vivo imaging, in vitro methods of organoid and microphysiological systems, as well as in silico quantitative physiological based pharmacokinetic and systems pharmacology models. Scientists continually leverage these advancements to interrogate the distribution of compounds in the CNS which may also show signals of substrate specificity of P-gp and/or BCRP. These methods have shown promise toward predicting and quantifying the unbound concentration(s) within the brain relevant for efficacy or safety. In this review, the authors have summarized the in vivo, in vitro, and proteomics advancements toward understanding the contribution of P-gp and/or BCRP in restricting the entry of compounds to the CNS of either healthy or special populations. Special emphasis has been provided on recent investigations on the application of a proteomics-informed approach to predict steady-state drug concentrations in the brain. Moreover, future perspectives regarding the role of these transporters in newer modalities are discussed.
Collapse
Affiliation(s)
- Sagnik Chatterjee
- Drug Metabolism and Pharmacokinetics, Ferring Pharmaceuticals A/S, Kastrup, Denmark
| | - Anup Arunrao Deshpande
- Drug Metabolism and Pharmacokinetics, Biocon Bristol Myers Squibb R&D Centre (BBRC), Syngene International Ltd, Bangalore, India
| | - Hong Shen
- Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Company, Princeton, New Jersey, USA
| |
Collapse
|
6
|
[11C]glyburide PET imaging for quantitative determination of the importance of Organic Anion-Transporting Polypeptide transporter function in the human liver and whole-body. Biomed Pharmacother 2022; 156:113994. [DOI: 10.1016/j.biopha.2022.113994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
|
7
|
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]
|
8
|
Breuil L, Goutal S, Marie S, Del Vecchio A, Audisio D, Soyer A, Goislard M, Saba W, Tournier N, Caillé F. Comparison of the Blood-Brain Barrier Transport and Vulnerability to P-Glycoprotein-Mediated Drug-Drug Interaction of Domperidone versus Metoclopramide Assessed Using In Vitro Assay and PET Imaging. Pharmaceutics 2022; 14:pharmaceutics14081658. [PMID: 36015284 PMCID: PMC9412994 DOI: 10.3390/pharmaceutics14081658] [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: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Domperidone and metoclopramide are widely prescribed antiemetic drugs with distinct neurological side effects. The impact of P-glycoprotein (P-gp)-mediated efflux at the blood−brain barrier (BBB) on brain exposure and BBB permeation was compared in vitro and in vivo using positron emission tomography (PET) imaging in rats with the radiolabeled analogs [11C]domperidone and [11C]metoclopramide. In P-gp-overexpressing cells, the IC50 of tariquidar, a potent P-gp inhibitor, was drastically different using [11C]domperidone (221 nM [198−248 nM]) or [11C]metoclopramide (4 nM [2−8 nM]) as the substrate. Complete P-gp inhibition led to a 1.8-fold higher increase in the cellular uptake of [11C]domperidone compared with [11C]metoclopramide (p < 0.0001). Brain PET imaging revealed that the baseline brain exposure (AUCbrain) of [11C]metoclopramide was 2.4-fold higher compared with [11C]domperidone (p < 0.001), consistent with a 1.8-fold higher BBB penetration (AUCbrain/AUCplasma). The maximal increase in the brain exposure (2.9-fold, p < 0.0001) and BBB penetration (2.9-fold, p < 0.0001) of [11C]metoclopramide was achieved using 8 mg/kg of tariquidar. In comparison, neither 8 nor 15 mg/kg of tariquidar increased the brain exposure of [11C]domperidone (p > 0.05). Domperidone is an avid P-gp substrate that was in vitro compared with metoclopramide. Domperidone benefits from a lower brain exposure and a limited risk for P-gp-mediated drug−drug interaction involving P-gp inhibition at the BBB.
Collapse
Affiliation(s)
- Louise Breuil
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
- Pharmacy Department, Robert-Debré Hospital, AP-HP, Université Paris Cité, 75019 Paris, France
| | - Sébastien Goutal
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
| | - Solène Marie
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
- Pharmacy Department, Bicêtre Hospital, AP-HP, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Antonio Del Vecchio
- CEA, Département Médicaments et Technologies pour la Santé, SCBM, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Davide Audisio
- CEA, Département Médicaments et Technologies pour la Santé, SCBM, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Amélie Soyer
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
| | - Maud Goislard
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
| | - Wadad Saba
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
- Correspondence:
| | - Fabien Caillé
- Laboratoire d’Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, 4 place du Général Leclerc, 91401 Orsay, France
| |
Collapse
|
9
|
Marie S, Hernández-Lozano I, Le Vée M, Breuil L, Saba W, Goislard M, Goutal S, Truillet C, Langer O, Fardel O, Tournier N. Pharmacokinetic Imaging Using 99mTc-Mebrofenin to Untangle the Pattern of Hepatocyte Transporter Disruptions Induced by Endotoxemia in Rats. Pharmaceuticals (Basel) 2022; 15:ph15040392. [PMID: 35455390 PMCID: PMC9028474 DOI: 10.3390/ph15040392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/09/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
Endotoxemia-induced inflammation may impact the activity of hepatocyte transporters, which control the hepatobiliary elimination of drugs and bile acids. 99mTc-mebrofenin is a non-metabolized substrate of transporters expressed at the different poles of hepatocytes. 99mTc-mebrofenin imaging was performed in rats after the injection of lipopolysaccharide (LPS). Changes in transporter expression were assessed using quantitative polymerase chain reaction of resected liver samples. Moreover, the particular impact of pharmacokinetic drug–drug interactions in the context of endotoxemia was investigated using rifampicin (40 mg/kg), a potent inhibitor of hepatocyte transporters. LPS increased 99mTc-mebrofenin exposure in the liver (1.7 ± 0.4-fold). Kinetic modeling revealed that endotoxemia did not impact the blood-to-liver uptake of 99mTc-mebrofenin, which is mediated by organic anion-transporting polypeptide (Oatp) transporters. However, liver-to-bile and liver-to-blood efflux rates were dramatically decreased, leading to liver accumulation. The transcriptomic profile of hepatocyte transporters consistently showed a downregulation of multidrug resistance-associated proteins 2 and 3 (Mrp2 and Mrp3), which mediate the canalicular and sinusoidal efflux of 99mTc-mebrofenin in hepatocytes, respectively. Rifampicin effectively blocked both the Oatp-mediated influx and the Mrp2/3-related efflux of 99mTc-mebrofenin. The additive impact of endotoxemia and rifampicin led to a 3.0 ± 1.3-fold increase in blood exposure compared with healthy non-treated animals. 99mTc-mebrofenin imaging is useful to investigate disease-associated change in hepatocyte transporter function.
Collapse
Affiliation(s)
- Solène Marie
- Université Paris-Saclay, CEA, CNRS, Inserm, Laboratoire d'Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
- Faculté de Pharmacie, Université Paris-Saclay, 92296 Châtenay-Malabry, France
- AP-HP, Université Paris-Saclay, Hôpital Bicêtre, Pharmacie Clinique, 94270 Le Kremlin Bicêtre, France
| | | | - Marc Le Vée
- Univ. Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, 35043 Rennes, France
| | - Louise Breuil
- Université Paris-Saclay, CEA, CNRS, Inserm, Laboratoire d'Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| | - Wadad Saba
- Université Paris-Saclay, CEA, CNRS, Inserm, Laboratoire d'Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| | - Maud Goislard
- Université Paris-Saclay, CEA, CNRS, Inserm, Laboratoire d'Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| | - Sébastien Goutal
- Université Paris-Saclay, CEA, CNRS, Inserm, Laboratoire d'Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, Laboratoire d'Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Olivier Fardel
- Univ. Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, 35043 Rennes, France
| | - Nicolas Tournier
- Université Paris-Saclay, CEA, CNRS, Inserm, Laboratoire d'Imagerie Biomédicale Multimodale, BIOMAPS, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| |
Collapse
|
10
|
Pastor CM, Brouwer KLR. New Pharmacokinetic Parameters of Imaging Substrates Quantified from Rat Liver Compartments. Drug Metab Dispos 2022; 50:58-64. [PMID: 34670777 DOI: 10.1124/dmd.121.000546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022] Open
Abstract
Hepatobiliary imaging is increasingly used by pharmacologists to quantify liver concentrations of transporter-dependent drugs. However, liver imaging does not quantify concentrations in extracellular space, hepatocytes, and bile canaliculi. Our study compared the compartmental distribution of two hepatobiliary substrates gadobenate dimeglumine [BOPTA; 0.08 liver extraction ratio (ER)] and mebrofenin (MEB; 0.93 ER) in a model of perfused rat liver. A gamma counter placed over livers measured liver concentrations. Livers were preperfused with gadopentetate dimeglumine to measure extracellular concentrations. Concentrations coming from bile canaliculi and hepatocytes were calculated. Transporter activities were assessed by concentration ratios between compartments and pharmacokinetic parameters that describe the accumulation and decay profiles of hepatocyte concentrations. The high liver concentrations of MEB relied mainly on hepatocyte and bile canaliculi concentrations. In contrast, the three compartments contributed to the low liver concentrations obtained during BOPTA perfusion. Nonlinear regression analysis of substrate accumulation in hepatocytes revealed that cellular efflux is measurable ∼4 minutes after the start of perfusion. The hepatocyte-to-extracellular concentration ratio measured at this time point was much higher during MEB perfusion. BOPTA transport by multidrug resistance associated protein 2 induced an aquaporin-mediated water transport, whereas MEB transport did not. BOPTA clearance from hepatocytes to bile canaliculi was higher than MEB clearance. MEB did not efflux back to sinusoids, whereas BOPTA basolateral efflux contributed to the decrease in hepatocyte concentrations. In conclusion, our ex vivo model quantifies substrate compartmental distribution and transport across hepatocyte membranes and provides an additional understanding of substrate distribution in the liver. SIGNIFICANCE STATEMENT: When transporter-dependent drugs target hepatocytes, cellular concentrations are important to investigate. Low concentrations on cellular targets impair drug therapeutic effects, whereas excessive hepatocyte concentrations may induce cellular toxicity. With a gamma counter placed over rat perfused livers, we measured substrate concentrations in the extracellular space, hepatocytes, and bile canaliculi. Transport across hepatocyte membranes was calculated. The study provides an additional understanding of substrate distribution in the liver.
Collapse
Affiliation(s)
- Catherine M Pastor
- Department of Radiology, University Hospital of Geneva, Switzerland (C.M.P.); Université de Paris, Centre de recherche sur l'inflammation, Inserm, U1149, CNRS, ERL8252, F-75006 Paris, France (C.M.P.); and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (K.L.R.B.)
| | - Kim L R Brouwer
- Department of Radiology, University Hospital of Geneva, Switzerland (C.M.P.); Université de Paris, Centre de recherche sur l'inflammation, Inserm, U1149, CNRS, ERL8252, F-75006 Paris, France (C.M.P.); and Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (K.L.R.B.)
| |
Collapse
|
11
|
Breuil L, Marie S, Goutal S, Auvity S, Truillet C, Saba W, Langer O, Caillé F, Tournier N. Comparative vulnerability of PET radioligands to partial inhibition of P-glycoprotein at the blood-brain barrier: A criterion of choice? J Cereb Blood Flow Metab 2022; 42:175-185. [PMID: 34496661 PMCID: PMC8721783 DOI: 10.1177/0271678x211045444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Only partial deficiency/inhibition of P-glycoprotein (P-gp, ABCB1) function at the blood-brain barrier (BBB) is likely to occur in pathophysiological situations or drug-drug interactions. This raises questions regarding the sensitivity of available PET imaging probes to detect moderate changes in P-gp function at the living BBB. In vitro, the half-maximum inhibitory concentration (IC50) of the potent P-gp inhibitor tariquidar in P-gp-overexpressing cells was significantly different using either [11C]verapamil (44 nM), [11C]N-desmethyl-loperamide (19 nM) or [11C]metoclopramide (4 nM) as substrate probes. In vivo PET imaging in rats showed that the half-maximum inhibition of P-gp-mediated efflux of [11C]metoclopramide, achieved using 1 mg/kg tariquidar (in vivo IC50 = 82 nM in plasma), increased brain exposure by 2.1-fold for [11C]metoclopramide (p < 0.05, n = 4) and 2.4-fold for [11C]verapamil (p < 0.05, n = 4), whereby cerebral uptake of the "avid" substrate [11C]N-desmethyl-loperamide was unaffected (p > 0.05, n = 4). This comparative study points to differences in the "vulnerability" to P-gp inhibition among radiolabeled substrates, which were apparently unrelated to their "avidity" (maximal response to P-gp inhibition). Herein, we advocate that partial inhibition of transporter function, in addition to complete inhibition, should be a primary criterion of evaluation regarding the sensitivity of radiolabeled substrates to detect moderate but physiologically-relevant changes in transporter function in vivo.
Collapse
Affiliation(s)
- Louise Breuil
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay France.,Pharmacy Department, Robert-Debré Hospital, AP-HP, Université de Paris, Paris, France
| | - Solène Marie
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay France.,Pharmacy Department, Bicêtre Hospital, AP-HP, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Sébastien Goutal
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay France
| | - Sylvain Auvity
- Pharmacy Department, Necker Hospital, AP-HP, UMR-S 1144, Université de Paris, Paris, France
| | - Charles Truillet
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay France
| | - Wadad Saba
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay France
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Fabien Caillé
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay France
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay France
| |
Collapse
|
12
|
Hernández-Lozano I, Mairinger S, Filip T, Sauberer M, Wanek T, Stanek J, Sake JA, Pekar T, Ehrhardt C, Langer O. PET imaging to assess the impact of P-glycoprotein on pulmonary drug delivery in rats. J Control Release 2021; 342:44-52. [PMID: 34971693 DOI: 10.1016/j.jconrel.2021.12.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/26/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
Several drugs approved for inhalation for the treatment of pulmonary diseases are substrates of the adenosine triphosphate-binding cassette (ABC) transporter P-glycoprotein (P-gp). P-gp is expressed in the apical membrane of pulmonary epithelial cells and could play a role in modulating the pulmonary absorption and distribution of inhaled drugs, thereby potentially contributing to variability in therapeutic response and/or systemic side effects. We developed a new in vivo experimental approach to assess the functional impact of P-gp on the pulmonary delivery of inhaled drugs in rats. By using positron emission tomography (PET) imaging, we measured the intrapulmonary pharmacokinetics of the model P-gp substrates (R)-[11C]verapamil ([11C]VPM) and [11C]-N-desmethyl-loperamide ([11C]dLOP) administered by intratracheal aerosolization in three rat groups: wild-type, Abcb1a/b(-/-) and wild-type treated with the P-gp inhibitor tariquidar. Lung exposure (AUClung_right) to [11C]VPM was 64% and 50% lower (p < 0.05) in tariquidar-treated and in Abcb1a/b(-/-) rats, respectively, compared to untreated wild-type rats. For [11C]dLOP, AUClung_right was 59% and 34% lower (p < 0.05) in tariquidar-treated and in Abcb1a/b(-/-) rats, respectively. Our results show that P-gp can affect the pulmonary disposition of inhaled P-gp substrates, whereby a decrease in P-gp activity may lead to lower lung exposure and potentially to a decrease in therapeutic efficacy. Our study highlights the potential of PET imaging with intratracheally aerosolized radiotracers to assess the impact of membrane transporters on pulmonary drug delivery, in rodents and potentially also in humans.
Collapse
Affiliation(s)
| | - Severin Mairinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of 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; Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria.
| |
Collapse
|
13
|
Tournier N, Comtat C, Lebon V, Gennisson JL. Challenges and Perspectives of the Hybridization of PET with Functional MRI or Ultrasound for Neuroimaging. Neuroscience 2021; 474:80-93. [DOI: 10.1016/j.neuroscience.2020.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
|
14
|
Kell DB. The Transporter-Mediated Cellular Uptake and Efflux of Pharmaceutical Drugs and Biotechnology Products: How and Why Phospholipid Bilayer Transport Is Negligible in Real Biomembranes. Molecules 2021; 26:5629. [PMID: 34577099 PMCID: PMC8470029 DOI: 10.3390/molecules26185629] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Over the years, my colleagues and I have come to realise that the likelihood of pharmaceutical drugs being able to diffuse through whatever unhindered phospholipid bilayer may exist in intact biological membranes in vivo is vanishingly low. This is because (i) most real biomembranes are mostly protein, not lipid, (ii) unlike purely lipid bilayers that can form transient aqueous channels, the high concentrations of proteins serve to stop such activity, (iii) natural evolution long ago selected against transport methods that just let any undesirable products enter a cell, (iv) transporters have now been identified for all kinds of molecules (even water) that were once thought not to require them, (v) many experiments show a massive variation in the uptake of drugs between different cells, tissues, and organisms, that cannot be explained if lipid bilayer transport is significant or if efflux were the only differentiator, and (vi) many experiments that manipulate the expression level of individual transporters as an independent variable demonstrate their role in drug and nutrient uptake (including in cytotoxicity or adverse drug reactions). This makes such transporters valuable both as a means of targeting drugs (not least anti-infectives) to selected cells or tissues and also as drug targets. The same considerations apply to the exploitation of substrate uptake and product efflux transporters in biotechnology. We are also beginning to recognise that transporters are more promiscuous, and antiporter activity is much more widespread, than had been realised, and that such processes are adaptive (i.e., were selected by natural evolution). The purpose of the present review is to summarise the above, and to rehearse and update readers on recent developments. These developments lead us to retain and indeed to strengthen our contention that for transmembrane pharmaceutical drug transport "phospholipid bilayer transport is negligible".
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK;
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs Lyngby, Denmark
- Mellizyme Biotechnology Ltd., IC1, Liverpool Science Park, Mount Pleasant, Liverpool L3 5TF, UK
| |
Collapse
|
15
|
Hanafy AS, Dietrich D, Fricker G, Lamprecht A. Blood-brain barrier models: Rationale for selection. Adv Drug Deliv Rev 2021; 176:113859. [PMID: 34246710 DOI: 10.1016/j.addr.2021.113859] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 01/21/2023]
Abstract
Brain delivery is a broad research area, the outcomes of which are far hindered by the limited permeability of the blood-brain barrier (BBB). Over the last century, research has been revealing the BBB complexity and the crosstalk between its cellular and molecular components. Pathologically, BBB alterations may precede as well as be concomitant or lead to brain diseases. To simulate the BBB and investigate options for drug delivery, several in vitro, in vivo, ex vivo, in situ and in silico models are used. Hundreds of drug delivery vehicles successfully pass preclinical trials but fail in clinical settings. Inadequate selection of BBB models is believed to remarkably impact the data reliability leading to unsatisfactory results in clinical trials. In this review, we suggest a rationale for BBB model selection with respect to the addressed research question and downstream applications. The essential considerations of an optimal BBB model are discussed.
Collapse
Affiliation(s)
- Amira Sayed Hanafy
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Dirk Dietrich
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany.
| |
Collapse
|
16
|
Hernández-Lozano I, Mairinger S, Traxl A, Sauberer M, Filip T, Stanek J, Kuntner C, Wanek T, Langer O. Assessing the Functional Redundancy between P-gp and BCRP in Controlling the Brain Distribution and Biliary Excretion of Dual Substrates with PET Imaging in Mice. Pharmaceutics 2021; 13:1286. [PMID: 34452247 PMCID: PMC8399697 DOI: 10.3390/pharmaceutics13081286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/20/2022] Open
Abstract
P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) are co-localized at the blood-brain barrier, where they display functional redundancy to restrict the brain distribution of dual P-gp/BCRP substrate drugs. We used positron emission tomography (PET) with the metabolically stable P-gp/BCRP substrates [11C]tariquidar, [11C]erlotinib, and [11C]elacridar to assess whether a similar functional redundancy as at the BBB exists in the liver, where both transporters mediate the biliary excretion of drugs. Wild-type, Abcb1a/b(-/-), Abcg2(-/-), and Abcb1a/b(-/-)Abcg2(-/-) mice underwent dynamic whole-body PET scans after i.v. injection of either [11C]tariquidar, [11C]erlotinib, or [11C]elacridar. Brain uptake of all three radiotracers was markedly higher in Abcb1a/b(-/-)Abcg2(-/-) mice than in wild-type mice, while only moderately changed in Abcb1a/b(-/-) and Abcg2(-/-) mice. The transfer of radioactivity from liver to excreted bile was significantly lower in Abcb1a/b(-/-)Abcg2(-/-) mice and almost unchanged in Abcb1a/b(-/-) and Abcg2(-/-) mice (with the exception of [11C]erlotinib, for which biliary excretion was also significantly reduced in Abcg2(-/-) mice). Our data provide evidence for redundancy between P-gp and BCRP in controlling both the brain distribution and biliary excretion of dual P-gp/BCRP substrates and highlight the utility of PET as an upcoming tool to assess the effect of transporters on drug disposition at a whole-body level.
Collapse
Affiliation(s)
| | - Severin Mairinger
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
| | - Alexander Traxl
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
- Center of Biomedical Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Claudia Kuntner
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
17
|
Tran V, Lux F, Tournier N, Jego B, Maître X, Anisorac M, Comtat C, Jan S, Selmeczi K, Evans MJ, Tillement O, Kuhnast B, Truillet C. Quantitative Tissue Pharmacokinetics and EPR Effect of AGuIX Nanoparticles: A Multimodal Imaging Study in an Orthotopic Glioblastoma Rat Model and Healthy Macaque. Adv Healthc Mater 2021; 10:e2100656. [PMID: 34212539 DOI: 10.1002/adhm.202100656] [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: 04/06/2021] [Revised: 05/29/2021] [Indexed: 01/10/2023]
Abstract
AGuIX are emerging radiosensitizing nanoparticles (NPs) for precision radiotherapy (RT) under clinical evaluation (Phase 2). Despite being accompanied by MRI thanks to the presence of gadolinium (Gd) at its surface, more sensitive and quantifiable imaging technique should further leverage the full potential of this technology. In this study, it is shown that 89 Zr can be labeled on such NPs directly for positron emission tomography (PET) imaging with a simple and scalable method. The stability of such complexes is remarkable in vitro and in vivo. Using a glioblastoma orthotopic rat model, it is shown that injected 89 Zr-AGuIX is detectable inside the tumor for at least 1 week. Interestingly, the particles seem to efficiently infiltrate the tumor even in necrotic areas, which places great hope for the treatment of radioresistant tumor. Lastly, the first PET/MR whole-body imaging is performed in non-human primate (NHP), which further demonstrates the translational potential of these bimodal NP.
Collapse
Affiliation(s)
- Vu‐Long Tran
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - François Lux
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
- Institut Universitaire de France (IUF) Paris France
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Benoit Jego
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Xavier Maître
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Claude Comtat
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Sébastien Jan
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Michael J. Evans
- Department of Radiology and Biomedical Imaging University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Department of Pharmaceutical Chemistry University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Helen Diller Family Comprehensive Cancer Center University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
| | - Olivier Tillement
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
| | - Bertrand Kuhnast
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Charles Truillet
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| |
Collapse
|
18
|
Hernández-Lozano I, Wanek T, Sauberer M, Filip T, Mairinger S, Stanek J, Traxl A, Karch R, Schuetz JD, Langer O. Influence of ABC transporters on the excretion of ciprofloxacin assessed with PET imaging in mice. Eur J Pharm Sci 2021; 163:105854. [PMID: 33865975 DOI: 10.1016/j.ejps.2021.105854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/31/2022]
Abstract
Ciprofloxacin is a commonly prescribed fluoroquinolone antibiotic which is cleared by active tubular secretion and intestinal excretion. Ciprofloxacin is a known substrate of the ATP-binding cassette (ABC) transporters breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 4 (MRP4). In this work, we used positron emission tomography (PET) imaging to investigate the influence of BCRP, MRP4, MRP2 and P-glycoprotein (P-gp) on the excretion of [18F]ciprofloxacin in mice. Dynamic 90-min PET scans were performed after intravenous injection of [18F]ciprofloxacin in wild-type mice without and with pre-treatment with the broad-spectrum MRP inhibitor MK571. Moreover, [18F]ciprofloxacin PET scans were performed in Abcc4(-/-), Abcc2(-/-), Abcc4(-/-)Abcg2(-/-) and Abcb1a/b(-/-)Abcg2(-/-) mice. In addition to non-compartmental pharmacokinetic (PK) analysis, a novel three-compartment PK model was developed for a detailed assessment of the renal disposition of [18F]ciprofloxacin. In MK571 pre-treated mice, a significant increase in the blood exposure to [18F]ciprofloxacin was observed along with a significant reduction in the renal and intestinal clearances. PK modelling revealed a significant reduction in renal radioactivity uptake (CL1) and in the rate constants for transfer of radioactivity from the corticomedullary renal region into blood (k2) and urine (k3), respectively, after MK571 administration. No changes in the renal clearance or in the estimated kidney PK model parameters were observed in any of the studied knockout models, while a significant reduction in the intestinal clearance was observed in Abcc2(-/-) and Abcc4(-/-)Abcg2(-/-) mice. Our data failed to reveal a role of any of the studied ABC transporters in the tubular secretion of ciprofloxacin. This may indicate that ciprofloxacin is handled in the kidneys by more than one transporter family, most likely with a great degree of mutual functional redundancy. Our study highlights the potential of PET imaging for an assessment of transporter-mediated renal excretion of radiolabelled drugs.
Collapse
Affiliation(s)
- Irene Hernández-Lozano
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria.
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria.
| | - Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria.
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria.
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria.
| | - Alexander Traxl
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria.
| | - Rudolf Karch
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, 1090 Vienna, Austria.
| | - John D Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 38105 Memphis, TN, USA.
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
19
|
Marie S, Hernández-Lozano I, Langer O, Tournier N. Repurposing 99mTc-Mebrofenin as a Probe for Molecular Imaging of Hepatocyte Transporters. J Nucl Med 2021; 62:1043-1047. [PMID: 33674399 DOI: 10.2967/jnumed.120.261321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/01/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocyte transporters control the hepatobiliary elimination of many drugs, metabolites, and endogenous substances. Hepatocyte transporter function is altered in several pathophysiologic situations and can be modulated by certain drugs, with a potential impact for pharmacokinetics and drug-induced liver injury. The development of substrate probes with optimal properties for selective and quantitative imaging of hepatic transporters remains a challenge. 99mTc-mebrofenin has been used for decades for hepatobiliary scintigraphy, but the specific transporters controlling its liver kinetics have not been characterized until recently. These include sinusoidal influx transporters (organic anion-transporting polypeptides) responsible for hepatic uptake of 99mTc-mebrofenin, and efflux transporters (multidrug resistance-associated proteins) mediating its canalicular (liver-to-bile) and sinusoidal (liver-to-blood) excretion. Pharmacokinetic modeling enables molecular interpretation of 99mTc-mebrofenin scintigraphy data, thus offering a widely available translational method to investigate transporter-mediated drug-drug interactions in vivo. 99mTc-mebrofenin allows for phenotyping transporter function at the different poles of hepatocytes as a biomarker of liver function.
Collapse
Affiliation(s)
| | | | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale, BioMaps, Université Paris-Saclay, CEA, CNRS, INSERM, Service Hospitalier Frédéric Joliot, Orsay, France
| |
Collapse
|
20
|
Wang D, Chen F, Han Z, Yin Z, Ge X, Lei P. Relationship Between Amyloid-β Deposition and Blood-Brain Barrier Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2021; 15:695479. [PMID: 34349624 PMCID: PMC8326917 DOI: 10.3389/fncel.2021.695479] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloid-β (Aβ) is the predominant pathologic protein in Alzheimer's disease (AD). The production and deposition of Aβ are important factors affecting AD progression and prognosis. The deposition of neurotoxic Aβ contributes to damage of the blood-brain barrier. However, the BBB is also crucial in maintaining the normal metabolism of Aβ, and dysfunction of the BBB aggravates Aβ deposition. This review characterizes Aβ deposition and BBB damage in AD, summarizes their interactions, and details their respective mechanisms.
Collapse
Affiliation(s)
- Dong Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | | | - Zhaoli Han
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Zhenyu Yin
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Xintong Ge
- Tianjin Neurological Institute, Tianjin, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
21
|
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
|
22
|
El Biali M, Karch R, Philippe C, Haslacher H, Tournier N, Hacker M, Zeitlinger M, Schmidl D, Langer O, Bauer M. ABCB1 and ABCG2 Together Limit the Distribution of ABCB1/ABCG2 Substrates to the Human Retina and the ABCG2 Single Nucleotide Polymorphism Q141K (c.421C> A) May Lead to Increased Drug Exposure. Front Pharmacol 2021; 12:698966. [PMID: 34220523 PMCID: PMC8242189 DOI: 10.3389/fphar.2021.698966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/04/2021] [Indexed: 12/26/2022] Open
Abstract
The widely expressed and poly-specific ABC transporters breast cancer resistance protein (ABCG2) and P-glycoprotein (ABCB1) are co-localized at the blood-brain barrier (BBB) and have shown to limit the brain distribution of several clinically used ABCB1/ABCG2 substrate drugs. It is currently not known to which extent these transporters, which are also expressed at the blood-retinal barrier (BRB), may limit drug distribution to the human eye and whether the ABCG2 reduced-function single-nucleotide polymorphism (SNP) Q141K (c.421C > A) has an impact on retinal drug distribution. Ten healthy male volunteers (five subjects with the c.421CC and c.421CA genotype, respectively) underwent two consecutive positron emission tomography (PET) scans after intravenous injection of the model ABCB1/ABCG2 substrate [11C]tariquidar. The second PET scan was performed with concurrent intravenous infusion of unlabelled tariquidar to inhibit ABCB1 in order to specifically reveal ABCG2 function.In response to ABCB1 inhibition with unlabelled tariquidar, ABCG2 c.421C > A genotype carriers showed significant increases (as compared to the baseline scan) in retinal radiotracer influx K 1 (+62 ± 57%, p = 0.043) and volume of distribution V T (+86 ± 131%, p = 0.043), but no significant changes were observed in subjects with the c.421C > C genotype. Our results provide the first evidence that ABCB1 and ABCG2 may together limit the distribution of systemically administered ABCB1/ABCG2 substrate drugs to the human retina. Functional redundancy between ABCB1 and ABCG2 appears to be compromised in carriers of the c.421C > A SNP who may therefore be more susceptible to transporter-mediated drug-drug interactions at the BRB than non-carriers.
Collapse
Affiliation(s)
- Myriam El Biali
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, VIE, Austria
| | - Rudolf Karch
- Centre for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, VIE, Austria
| | - Cécile Philippe
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, VIE, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, VIE, Austria
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, VIE, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, VIE, Austria
| | - Doreen Schmidl
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, VIE, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, VIE, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, VIE, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, VIE, Austria
| |
Collapse
|
23
|
Pharmacokinetic neuroimaging to study the dose-related brain kinetics and target engagement of buprenorphine in vivo. Neuropsychopharmacology 2021; 46:1220-1228. [PMID: 33603137 PMCID: PMC8115308 DOI: 10.1038/s41386-021-00976-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/18/2020] [Accepted: 01/24/2021] [Indexed: 12/31/2022]
Abstract
A wide range of buprenorphine doses are used for either pain management or maintenance therapy in opioid addiction. The complex in vitro profile of buprenorphine, with affinity for µ-, δ-, and κ-opioid receptors (OR), makes it difficult to predict its dose-related neuropharmacology in vivo. In rats, microPET imaging and pretreatment by OR antagonists were performed to assess the binding of radiolabeled buprenorphine (microdose 11C-buprenorphine) to OR subtypes in vivo (n = 4 per condition). The µ-selective antagonist naloxonazine (10 mg/kg) and the non-selective OR antagonist naloxone (1 mg/kg) blocked the binding of 11C-buprenorphine, while pretreatment by the δ-selective (naltrindole, 3 mg/kg) or the κ-selective antagonist (norbinaltorphimine, 10 mg/kg) did not. In four macaques, PET imaging and kinetic modeling enabled description of the regional brain kinetics of 11C-buprenorphine, co-injected with increasing doses of unlabeled buprenorphine. No saturation of the brain penetration of buprenorphine was observed for doses up to 0.11 mg/kg. Regional differences in buprenorphine-associated receptor occupancy were observed. Analgesic doses of buprenorphine (0.003 and 0.006 mg/kg), respectively, occupied 20% and 49% of receptors in the thalamus while saturating the low but significant binding observed in cerebellum and occipital cortex. Occupancy >90% was achieved in most brain regions with plasma concentrations >7 µg/L. PET data obtained after co-injection of an analgesic dose of buprenorphine (0.003 mg/kg) predicted the binding potential of microdose 11C-buprenorphine. This strategy could be further combined with pharmacodynamic exploration or pharmacological MRI to investigate the neuropharmacokinetics and neuroreceptor correlate, at least at µ-OR, of the acute effects of buprenorphine in humans.
Collapse
|
24
|
Williamson B, Pilla Reddy V. Blood retinal barrier and ocular pharmacokinetics: Considerations for the development of oncology drugs. Biopharm Drug Dispos 2021; 42:128-136. [PMID: 33759216 DOI: 10.1002/bdd.2276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/09/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are an example of targeted drug therapy to treat cancer while minimizing damage to healthy tissue. In contrast to traditional oncology drugs, the toxicity profile of targeted therapies is less well understood and can include severe ocular adverse events, which are among the most common toxicity reported by these therapeutics. Inhibition of Mer receptor tyrosine kinase (MERTK) promotes innate tumor immunity by decreasing M2-macrophage polarization and efferocytosis. This mechanism offers the opportunity for targeted immunotherapy to treat cancer; however, the ocular expression of MERTK increases the difficulty for developing a targeted drug due to toxicity concerns. In this article we review the pharmacokinetic (PK) parameters and in vitro absorption, distribution, metabolism, and excretion (ADME) assays available to evaluate ocular disposition and assess the relationship between clinical PK and reported ocular events for TKIs to allow backtranslation to preclinical models. Understanding the ocular disposition in the context of PK and safety remains an evolving area and is likely to be a key aspect of developing safe and efficacious oncology drugs, devoid of ocular toxicity.
Collapse
Affiliation(s)
- Beth Williamson
- Drug Metabolism and Pharmacokinetics, Early Oncology, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Venkatesh Pilla Reddy
- Modelling and Simulation, Early Oncology, Oncology R&D, AstraZeneca, Cambridge, UK.,Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
25
|
Bauer M, Barna S, Blaickner M, Prosenz K, Bamminger K, Pichler V, Tournier N, Hacker M, Zeitlinger M, Karanikas G, Langer O. Human Biodistribution and Radiation Dosimetry of the P-Glycoprotein Radiotracer [ 11C]Metoclopramide. Mol Imaging Biol 2021; 23:180-185. [PMID: 33481175 PMCID: PMC7910245 DOI: 10.1007/s11307-021-01582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE To assess in healthy volunteers the whole-body distribution and dosimetry of [11C]metoclopramide, a new positron emission tomography (PET) tracer to measure P-glycoprotein activity at the blood-brain barrier. PROCEDURES Ten healthy volunteers (five women, five men) were intravenously injected with 387 ± 49 MBq of [11C]metoclopramide after low dose CT scans and were then imaged by whole-body PET scans from head to upper thigh over approximately 70 min. Ten source organs (brain, thyroid gland, right lung, myocardium, liver, gall bladder, left kidney, red bone marrow, muscle and the contents of the urinary bladder) were manually delineated on whole-body images. Absorbed doses were calculated with QDOSE (ABX-CRO) using the integrated IDAC-Dose 2.1 module. RESULTS The majority of the administered dose of [11C]metoclopramide was taken up into the liver followed by urinary excretion and, to a smaller extent, biliary excretion of radioactivity. The mean effective dose of [11C]metoclopramide was 1.69 ± 0.26 μSv/MBq for female subjects and 1.55 ± 0.07 μSv/MBq for male subjects. The two organs receiving the highest radiation doses were the urinary bladder (10.81 ± 0.23 μGy/MBq and 8.78 ± 0.89 μGy/MBq) and the liver (6.80 ± 0.78 μGy/MBq and 4.91 ± 0.74 μGy/MBq) for female and male subjects, respectively. CONCLUSIONS [11C]Metoclopramide showed predominantly renal excretion, and is safe and well tolerated in healthy adults. The effective dose of [11C]metoclopramide was comparable to other 11C-labeled PET tracers.
Collapse
Affiliation(s)
- Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.
| | - Sandra Barna
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Matthias Blaickner
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
- Center for Medical Physics Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Konstantin Prosenz
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Karsten Bamminger
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Verena Pichler
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Marcus Hacker
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Georgios Karanikas
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
26
|
Zhang Q, He Z, Liu Z, Gong L. Integrated plasma and liver gas chromatography mass spectrometry and liquid chromatography mass spectrometry metabolomics to reveal physiological functions of sodium taurocholate cotransporting polypeptide (NTCP) with an Ntcp knockout mouse model. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1165:122531. [DOI: 10.1016/j.jchromb.2021.122531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022]
|
27
|
Bourasset F, Auvity S, Thorne RG, Scherrmann JM. Brain Distribution of Drugs: Brain Morphology, Delivery Routes, and Species Differences. Handb Exp Pharmacol 2021; 273:97-120. [PMID: 33474672 DOI: 10.1007/164_2020_402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Neuropharmacokinetics considers cerebral drug distribution as a critical process for central nervous system drug action as well as for drug penetration through the CNS barriers. Brain distribution of small molecules obeys classical rules of drug partition, permeability, binding to fluid proteins or tissue components, and tissue perfusion. The biodistribution of all drugs, including both small molecules and biologics, may also be influenced by specific brain properties related to brain anatomy and physiological barriers, fluid dynamics, and cellular and biochemical composition, each of which can exhibit significant interspecies differences. All of these properties contribute to select optimal dosing paradigms and routes of drug delivery to reach brain targets for classical small molecule drugs as well as for biologics. The importance of these properties for brain delivery and exposure also highlights the need for efficient new analytical technologies to more comprehensively investigate drug distribution in the CNS, a complex multi-compartmentalized organ system.
Collapse
Affiliation(s)
- Fanchon Bourasset
- Faculty of Pharmacy, University of Paris, Paris, France.,INSERM UMR-S1144, Paris, France
| | - Sylvain Auvity
- Faculty of Pharmacy, University of Paris, Paris, France.,INSERM UMR-S1144, Paris, France
| | - Robert G Thorne
- Denali Therapeutics, Inc., South San Francisco, CA, USA. .,Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA.
| | - Jean-Michel Scherrmann
- Faculty of Pharmacy, University of Paris, Paris, France. .,INSERM UMR-S1144, Paris, France.
| |
Collapse
|
28
|
Stieger B, Steiger J, Locher KP. Membrane lipids and transporter function. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166079. [PMID: 33476785 DOI: 10.1016/j.bbadis.2021.166079] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/12/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Transport proteins are essential for cells in allowing the exchange of substances between cells and their environment across the lipid bilayer forming a tight barrier. Membrane lipids modulate the function of transmembrane proteins such as transporters in two ways: Lipids are tightly and specifically bound to transport proteins and in addition they modulate from the bulk of the lipid bilayer the function of transport proteins. This overview summarizes currently available information at the ultrastructural level on lipids tightly bound to transport proteins and the impact of altered bulk membrane lipid composition. Human diseases leading to altered lipid homeostasis will lead to altered membrane lipid composition, which in turn affect the function of transporter proteins.
Collapse
Affiliation(s)
- Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland.
| | - Julia Steiger
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Kaspar P Locher
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
29
|
Hernández-Lozano I, Mairinger S, Sauberer M, Stanek J, Filip T, Wanek T, Ciarimboli G, Tournier N, Langer O. Influence of Cation Transporters (OCTs and MATEs) on the Renal and Hepatobiliary Disposition of [ 11C]Metoclopramide in Mice. Pharm Res 2021; 38:127-140. [PMID: 33559045 PMCID: PMC7902338 DOI: 10.1007/s11095-021-03002-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/04/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE To investigate the role of cation transporters (OCTs, MATEs) in the renal and hepatic disposition of the radiolabeled antiemetic drug [11C]metoclopramide in mice with PET. METHODS PET was performed in wild-type mice after administration of an intravenous microdose (<1 μg) of [11C]metoclopramide without and with co-administration of either unlabeled metoclopramide (5 or 10 mg/kg) or the prototypical cation transporter inhibitors cimetidine (150 mg/kg) or sulpiride (25 mg/kg). [11C]Metoclopramide PET was also performed in wild-type and Slc22a1/2(-/-) mice. Radiolabeled metabolites were measured at 15 min after radiotracer injection and PET data were corrected for radiolabeled metabolites. RESULTS [11C]Metoclopramide was highly metabolized and [11C]metoclopramide-derived radioactivity was excreted into the urine. The different investigated treatments decreased (~2.5-fold) the uptake of [11C]metoclopramide from plasma into the kidney and liver, inhibited metabolism and decreased (up to 3.8-fold) urinary excretion, which resulted in increased plasma concentrations of [11C]metoclopramide. Kidney and liver uptake were moderately (~1.3-fold) reduced in Slc22a1/2(-/-) mice. CONCLUSIONS Our results suggest a contribution of OCT1/2 to the kidney and liver uptake and of MATEs to the urinary excretion of [11C]metoclopramide in mice. Cation transporters may contribute, next to variability in the activity of metabolizing enzymes, to variability in metoclopramide pharmacokinetics and side effects.
Collapse
Affiliation(s)
- Irene Hernández-Lozano
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Giuliano Ciarimboli
- Medicine Clinic D. Experimental Nephrology, University Hospital Münster, Münster, Germany
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - 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.
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
30
|
García-Varela L, García DV, Kakiuchi T, Ohba H, Nishiyama S, Tago T, Elsinga PH, Tsukada H, Colabufo NA, Dierckx RAJO, van Waarde A, Toyohara J, Boellaard R, Luurtsema G. Pharmacokinetic Modeling of ( R)-[ 11C]verapamil to Measure the P-Glycoprotein Function in Nonhuman Primates. Mol Pharm 2020; 18:416-428. [PMID: 33315404 PMCID: PMC7788571 DOI: 10.1021/acs.molpharmaceut.0c01014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
(R)-[11C]verapamil is a radiotracer
widely used for the evaluation of the P-glycoprotein (P-gp) function
at the blood–brain barrier (BBB). Several studies have evaluated
the pharmacokinetics of (R)-[11C]verapamil
in rats and humans under different conditions. However, to the best
of our knowledge, the pharmacokinetics of (R)-[11C]verapamil have not yet been evaluated in nonhuman primates.
Our study aims to establish (R)-[11C]verapamil
as a reference P-gp tracer for comparison of a newly developed P-gp
positron emission tomography (PET) tracer in a species close to humans.
Therefore, the study assesses the kinetics of (R)-[11C]verapamil and evaluates the effect of scan duration and
P-gp inhibition on estimated pharmacokinetic parameters. Three nonhuman
primates underwent two dynamic 91 min PET scans with arterial blood
sampling, one at baseline and another after inhibition of the P-gp
function. The (R)-[11C]verapamil data
were analyzed using 1-tissue compartment model (1-TCM) and 2-tissue
compartment model fits using plasma-corrected for polar radio-metabolites
or non-corrected for radio-metabolites as an input function and with
various scan durations (10, 20, 30, 60, and 91 min). The preferred
model was chosen according to the Akaike information criterion and
the standard errors (SE %) of the estimated parameters. 1-TCM was
selected as the model of choice to analyze the (R)-[11C]verapamil data at baseline and after inhibition
and for all scan durations tested. The volume of distribution (VT) and the efflux constant k2 estimations were affected by the evaluated scan durations,
whereas the influx constant K1 estimations
remained relatively constant. After P-gp inhibition (tariquidar, 8
mg/kg), in a 91 min scan duration, the whole-brain VT increased significantly up to 208% (p < 0.001) and K1 up to 159% (p < 0.001) compared with baseline scans. The k2 values decreased significantly after P-gp
inhibition in all the scan durations except for the 91 min scans.
This study suggests the use of K1, calculated
with 1-TCM and using short PET scans (10 to 30 min), as a suitable
parameter to measure the P-gp function at the BBB of nonhuman primates.
Collapse
Affiliation(s)
- Lara García-Varela
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, The Netherlands
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, The Netherlands
| | - Takeharu Kakiuchi
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu 434-8601, Shizuoka, Japan
| | - Hiroyuki Ohba
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu 434-8601, Shizuoka, Japan
| | - Shingo Nishiyama
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu 434-8601, Shizuoka, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, The Netherlands
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu 434-8601, Shizuoka, Japan
| | - Nicola A Colabufo
- Department of Pharmacy, University of Bari Aldo Moro, Bari 70125, Italy.,Biofordrug, Spin-off Università degli Studi di Bari "A. Moro", via Dante 99, Triggiano, Bari 70019, Italy
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, The Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, The Netherlands
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, The Netherlands
| |
Collapse
|
31
|
Samarehfekri H, Rahimi HR, Ranjbar M. Controlled and cellulose eco-friendly synthesis and characterization of Bi 2O 2CO 3 quantum dot nanostructures (QDNSs) and drug delivery study. Sci Rep 2020; 10:21302. [PMID: 33277600 PMCID: PMC7718884 DOI: 10.1038/s41598-020-78266-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
This work aimed to prepare solvent-free or green Bi2O2CO3 for quantum dot nanostructures (QDNSs) based on cellulose as a stabilizer and green capping agent to sorafenib delivery for liver targeting. Because the walnut tree is one of the most abundant trees in Iran, it was tried to synthesize Bi2O2CO3 QDNSs using a walnut skin extract. The saturation magnetization for Bi2O2CO3 QDNSs was calculated to be 68.1. Also, the size of products was measured at around 60–80 nm with the Debye–Scherrer equation. Moreover, the morphology, functional groups, and crystallography of the Bi2O2CO3 nanoparticles were investigated using atomic force microscopy, scanning electron microscopy, vibrating-sample magnetometer, and Uv–vis spectroscopy. The results demonstrated that Bi2O2CO3 QDNSs have opto-magnetic properties and they can be suggested as the candidate materials for the sorafenib delivery on the liver tissue. The optical band gap estimated for Bi2O2CO3 QDNSs was found to be red-shift from 3.22 eV. This study suggests the preparation of the Bi2O2CO3 QDNSs based on cellulose as new opto-magnetic materials at different temperatures of 180 °C, 200 °C, 220 °C, and 240 °C for sorafenib delivery as a type of biological therapy drug.
Collapse
Affiliation(s)
- Hojat Samarehfekri
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Reza Rahimi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Ranjbar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran. .,Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, P.O. Box: 76175-493, 76169-11319, Kerman, Iran.
| |
Collapse
|
32
|
Imagerie TEP pour l’étude des répercussions fonctionnelles de la P-glycoprotéine en neuropharmacocinétique. Therapie 2020; 75:623-632. [DOI: 10.1016/j.therap.2020.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/03/2019] [Accepted: 02/17/2020] [Indexed: 11/20/2022]
|
33
|
García-Varela L, Arif WM, Vállez García D, Kakiuchi T, Ohba H, Harada N, Tago T, Elsinga PH, Tsukada H, Colabufo NA, Dierckx RAJO, van Waarde A, Toyohara J, Boellaard R, Luurtsema G. Pharmacokinetic Modeling of [ 18F]MC225 for Quantification of the P-Glycoprotein Function at the Blood-Brain Barrier in Non-Human Primates with PET. Mol Pharm 2020; 17:3477-3486. [PMID: 32787277 PMCID: PMC7482398 DOI: 10.1021/acs.molpharmaceut.0c00514] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
![]()
[18F]MC225 has been developed as a weak substrate of
P-glycoprotein (P-gp) aimed to measure changes in the P-gp function
at the blood–brain barrier with positron emission tomography.
This study evaluates [18F]MC225 kinetics in non-human primates
and investigates the effect of both scan duration and P-gp inhibition.
Three rhesus monkeys underwent two 91-min dynamic scans with blood
sampling at baseline and after P-gp inhibition (8 mg/kg tariquidar).
Data were analyzed using the 1-tissue compartment model (1-TCM) and
2-tissue compartment model (2-TCM) fits using metabolite-corrected
plasma as the input function and for various scan durations (10, 20,
30, 60, and 91 min). The preferred model was chosen according to the
Akaike information criterion and the standard errors (%) of the estimated
parameters. For the 91-min scan duration, the influx constant K1 increased by 40.7% and the volume of distribution
(VT) by 30.4% after P-gp inhibition, while
the efflux constant k2 did not change
significantly. Similar changes were found for all evaluated scan durations. K1 did not depend on scan duration (10 min—K1 = 0.2191 vs 91 min—K1 = 0.2258), while VT and k2 did. A scan duration of 10 min seems sufficient
to properly evaluate the P-gp function using K1 obtained with 1-TCM. For the 91-min scan, VT and K1 can be estimated
with a 2-TCM, and both parameters can be used to assess P-gp function.
Collapse
Affiliation(s)
- Lara García-Varela
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands
| | - Wejdan M Arif
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands.,Department of Radiological Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands
| | - Takeharu Kakiuchi
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka 434-8601, Japan
| | - Hiroyuki Ohba
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka 434-8601, Japan
| | - Norihiro Harada
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka 434-8601, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics KK, 5000 Hirakuchi, Hamakita, Hamamatsu, Shizuoka 434-8601, Japan
| | - Nicola Antonio Colabufo
- Department of Pharmacy, University of Bari Aldo Moro, Bari 70121, Italy.,Biofordrug, Spin-off Università degli Studi di Bari "A. Moro", via Dante 99, Triggiano, Bari 70019, Italy
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, Groningen 9713 GZ, the Netherlands
| |
Collapse
|
34
|
Zoufal V, Mairinger S, Brackhan M, Krohn M, Filip T, Sauberer M, Stanek J, Wanek T, Tournier N, Bauer M, Pahnke J, Langer O. Imaging P-Glycoprotein Induction at the Blood-Brain Barrier of a β-Amyloidosis Mouse Model with 11C-Metoclopramide PET. J Nucl Med 2020; 61:1050-1057. [PMID: 31806767 PMCID: PMC7383073 DOI: 10.2967/jnumed.119.237198] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/18/2019] [Indexed: 01/30/2023] Open
Abstract
P-glycoprotein (ABC subfamily B member 1, ABCB1) plays an important role at the blood-brain barrier (BBB) in promoting clearance of neurotoxic β-amyloid (Aβ) peptides from the brain into the blood. ABCB1 expression and activity were found to be decreased in the brains of Alzheimer disease patients. Treatment with drugs that induce cerebral ABCB1 activity may be a promising approach to delay the build-up of Aβ deposits in the brain by enhancing clearance of Aβ peptides from the brain. The aim of this study was to investigate whether PET with the weak ABCB1 substrate radiotracer 11C-metoclopramide can measure ABCB1 induction at the BBB in a β-amyloidosis mouse model (APP/PS1-21 mice) and in wild-type mice. Methods: Groups of wild-type and APP/PS1-21 mice aged 50 or 170 d underwent 11C-metoclopramide baseline PET scans or scans after intraperitoneal treatment with the rodent pregnane X receptor activator 5-pregnen-3β-ol-20-one-16α-carbonitrile (PCN, 25 mg/kg) or its vehicle over 7 d. At the end of the PET scans, brains were harvested for immunohistochemical analysis of ABCB1 and Aβ levels. In separate groups of mice, radiolabeled metabolites of 11C-metoclopramide were determined in plasma and brain at 15 min after radiotracer injection. As an outcome parameter of cerebral ABCB1 activity, the elimination slope of radioactivity washout from the brain (kE,brain) was calculated. Results: PCN treatment resulted in an increased clearance of radioactivity from the brain as reflected by significant increases in kE,brain (from +26% to +54% relative to baseline). Immunohistochemical analysis confirmed ABCB1 induction in the brains of PCN-treated APP/PS1-21 mice with a concomitant decrease in Aβ levels. There was a significant positive correlation between kE,brain and ABCB1 levels in the brain. In wild-type mice, a significant age-related decrease in kE,brain was found. Metabolite analysis showed that most radioactivity in the brain comprised unmetabolized 11C-metoclopramide in all animal groups. Conclusion:11C-metoclopramide can measure ABCB1 induction in the mouse brain without the need to consider an arterial input function and may find potential application in Alzheimer disease patients to noninvasively evaluate strategies to enhance the clearance properties of the BBB.
Collapse
Affiliation(s)
- Viktoria Zoufal
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Mirjam Brackhan
- Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Markus Krohn
- Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Nicolas Tournier
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, INSERM, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
- LIED, University of Lübeck, Lübeck, Germany
- Leibniz-Institute of Plant Biochemistry, Halle, Germany
- Department of Pharmacology, Medical Faculty, University of Latvia, Rīga, Latvia; and
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging und Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Validation of Pharmacological Protocols for Targeted Inhibition of Canalicular MRP2 Activity in Hepatocytes Using [ 99mTc]mebrofenin Imaging in Rats. Pharmaceutics 2020; 12:pharmaceutics12060486. [PMID: 32471244 PMCID: PMC7355955 DOI: 10.3390/pharmaceutics12060486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022] Open
Abstract
The multidrug resistance-associated protein 2 (MRP2) mediates the biliary excretion of drugs and metabolites. [99mTc]mebrofenin may be employed as a probe for hepatic MRP2 activity because its biliary excretion is predominantly mediated by this transporter. As the liver uptake of [99mTc]mebrofenin depends on organic anion-transporting polypeptide (OATP) activity, a safe protocol for targeted inhibition of hepatic MRP2 is needed to study the intrinsic role of each transporter system. Diltiazem (DTZ) and cyclosporin A (CsA) were first confirmed to be potent MRP2 inhibitors in vitro. Dynamic acquisitions were performed in rats (n = 5-6 per group) to assess the kinetics of [99mTc]mebrofenin in the liver, intestine and heart-blood pool after increasing doses of inhibitors. Their impact on hepatic blood flow was assessed using Doppler ultrasound (n = 4). DTZ (s.c., 10 mg/kg) and low-dose CsA (i.v., 0.01 mg/kg) selectively decreased the transfer of [99mTc]mebrofenin from the liver to the bile (k3). Higher doses of DTZ and CsA did not further decrease k3 but dose-dependently decreased the uptake (k1) and backflux (k2) rate constants between blood and liver. High dose of DTZ (i.v., 3 mg/kg) but not CsA (i.v., 5 mg/kg) significantly decreased the blood flow in the portal vein and hepatic artery. Targeted pharmacological inhibition of hepatic MRP2 activity can be achieved in vivo without impacting OATP activity and liver blood flow. Clinical studies are warranted to validate [99mTc]mebrofenin in combination with low-dose CsA as a novel substrate/inhibitor pair to untangle the role of OATP and MRP2 activity in liver diseases.
Collapse
|
36
|
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]
|
37
|
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
|
38
|
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
|
39
|
Helms HCC, Kristensen M, Saaby L, Fricker G, Brodin B. Drug Delivery Strategies to Overcome the Blood-Brain Barrier (BBB). Handb Exp Pharmacol 2020; 273:151-183. [PMID: 33367937 DOI: 10.1007/164_2020_403] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The brain capillary endothelium serves both as an exchange site for gases and solutes between blood and brain and as a protective fence against neurotoxic compounds from the blood. While this "blood-brain barrier" (BBB) function protects the fragile environment in the brain, it also poses a tremendous challenge for the delivery of drug compounds to the brain parenchyma. Paracellular brain uptake of drug compounds is limited by the physical tightness of the endothelium, which is tightly sealed with junction complexes. Transcellular uptake of lipophilic drug compounds is limited by the activity of active efflux pumps in the luminal membrane. As a result, the majority of registered CNS drug compounds are small lipophilic compounds which are not efflux transporter substrates. Small molecule CNS drug development therefore focuses on identifying compounds with CNS target affinity and modifies these in order to optimize lipophilicity and decrease efflux pump interactions. Since efflux pump activity is limiting drug uptake, it has been investigated whether coadministration of drug compounds with efflux pump inhibitors could increase drug uptake. While the concept works to some extent, a lot of challenges have been encountered in terms of obtaining efficient inhibition while avoiding adverse effects.Some CNS drug compounds enter the brain via nutrient transport proteins, an example is the levodopa, a prodrug of Dopamine, which crosses the BBB via the large neutral amino acid transporter LAT1. While carrier-mediated transport of drug compounds may seem attractive, the development of drugs targeting transporters is very challenging, since the compounds should have a good fit to the binding site, while still maintaining their CNS target affinity.Receptor-mediated transport of drug compounds, especially biotherapeutics, conjugated to a receptor-binding ligand has shown some promise, although the amounts transported are rather low. This also holds true for drug-conjugation to cell-penetrating peptides. Due to the low uptake of biotherapeutics, barrier-breaching approaches such as mannitol injections and focused ultrasound have been employed with some success to patient groups with no other treatment options.
Collapse
Affiliation(s)
| | - Mie Kristensen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Saaby
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Bioneer-Farma, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Birger Brodin
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
40
|
Gomez-Zepeda D, Taghi M, Scherrmann JM, Decleves X, Menet MC. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019; 12:pharmaceutics12010020. [PMID: 31878061 PMCID: PMC7022905 DOI: 10.3390/pharmaceutics12010020] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Drug delivery into the brain is regulated by the blood-brain interfaces. The blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the blood-arachnoid barrier (BAB) regulate the exchange of substances between the blood and brain parenchyma. These selective barriers present a high impermeability to most substances, with the selective transport of nutrients and transporters preventing the entry and accumulation of possibly toxic molecules, comprising many therapeutic drugs. Transporters of the ATP-binding cassette (ABC) superfamily have an important role in drug delivery, because they extrude a broad molecular diversity of xenobiotics, including several anticancer drugs, preventing their entry into the brain. Gliomas are the most common primary tumors diagnosed in adults, which are often characterized by a poor prognosis, notably in the case of high-grade gliomas. Therapeutic treatments frequently fail due to the difficulty of delivering drugs through the brain barriers, adding to diverse mechanisms developed by the cancer, including the overexpression or expression de novo of ABC transporters in tumoral cells and/or in the endothelial cells forming the blood-brain tumor barrier (BBTB). Many models have been developed to study the phenotype, molecular characteristics, and function of the blood-brain interfaces as well as to evaluate drug permeability into the brain. These include in vitro, in vivo, and in silico models, which together can help us to better understand their implication in drug resistance and to develop new therapeutics or delivery strategies to improve the treatment of pathologies of the central nervous system (CNS). In this review, we present the principal characteristics of the blood-brain interfaces; then, we focus on the ABC transporters present on them and their implication in drug delivery; next, we present some of the most important models used for the study of drug transport; finally, we summarize the implication of ABC transporters in glioma and the BBTB in drug resistance and the strategies to improve the delivery of CNS anticancer drugs.
Collapse
Affiliation(s)
- David Gomez-Zepeda
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| | - Méryam Taghi
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Xavier Decleves
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Biologie du médicament et toxicologie, Hôpital Cochin, AP HP, 75006 Paris, France
| | - Marie-Claude Menet
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Hormonologie adulte, Hôpital Cochin, AP HP, 75006 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| |
Collapse
|
41
|
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
|
42
|
Roux GL, Jarray R, Guyot AC, Pavoni S, Costa N, Théodoro F, Nassor F, Pruvost A, Tournier N, Kiyan Y, Langer O, Yates F, Deslys JP, Mabondzo A. Proof-of-Concept Study of Drug Brain Permeability Between in Vivo Human Brain and an in Vitro iPSCs-Human Blood-Brain Barrier Model. Sci Rep 2019; 9:16310. [PMID: 31690750 PMCID: PMC6831611 DOI: 10.1038/s41598-019-52213-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022] Open
Abstract
The development of effective central nervous system (CNS) drugs has been hampered by the lack of robust strategies to mimic the blood-brain barrier (BBB) and cerebrovascular impairments in vitro. Recent technological advancements in BBB modeling using induced pluripotent stem cells (iPSCs) allowed to overcome some of these obstacles, nonetheless the pertinence for their use in drug permeation study remains to be established. This mandatory information requires a cross comparison of in vitro and in vivo pharmacokinetic data in the same species to avoid failure in late clinical drug development. Here, we measured the BBB permeabilities of 8 clinical positron emission tomography (PET) radioligands with known pharmacokinetic parameters in human brain in vivo with a newly developed in vitro iPSC-based human BBB (iPSC-hBBB) model. Our findings showed a good correlation between in vitro and in vivo drug brain permeability (R2 = 0.83; P = 0.008) which contrasted with the limited correlation between in vitro apparent permeability for a set of 18 CNS/non-CNS compounds using the in vitro iPSCs-hBBB model and drug physicochemical properties. Our data suggest that the iPSC-hBBB model can be integrated in a flow scheme of CNS drug screening and potentially used to study species differences in BBB permeation.
Collapse
Affiliation(s)
- Gwenaëlle Le Roux
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Rafika Jarray
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Anne-Cécile Guyot
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Serena Pavoni
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France
| | - Narciso Costa
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Frédéric Théodoro
- Service de Pharmacologie et d'Immunoanalyse (SPI), Plateforme Smart-MS, CEA, INRA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Ferid Nassor
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Alain Pruvost
- Service de Pharmacologie et d'Immunoanalyse (SPI), Plateforme Smart-MS, CEA, INRA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Nicolas Tournier
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Univ. Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Yulia Kiyan
- Medizinische Hochschule Hannover, DE-30625, Hannover, Germany
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria.,Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, A-2444, Seibersdorf, Austria
| | - Frank Yates
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Jean Philippe Deslys
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France
| | - Aloïse Mabondzo
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France.
| |
Collapse
|
43
|
Generation of a Small Library of Natural Products Designed to Cover Chemical Space Inexpensively. PHARMACEUTICAL FRONTIERS 2019; 1:e190005. [PMID: 31485581 PMCID: PMC6726486 DOI: 10.20900/pf20190005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural products space includes at least 200,000 compounds and the structures of most of these compounds are available in digital format. Previous analyses showed (i) that although they were capable of taking up synthetic pharmaceutical drugs, such exogenous molecules were likely the chief ‘natural’ substrates in the evolution of the transporters used to gain cellular entry by pharmaceutical drugs, and (ii) that a relatively simple but rapid clustering algorithm could produce clusters from which individual elements might serve to form a representative library covering natural products space. This exploited the fact that the larger clusters were likely to be formed early in evolution (and hence to have been accompanied by suitable transporters), so that very small clusters, including singletons, could be ignored. In the latter work, we assumed that the molecule chosen might be that in the middle of the cluster. However, this ignored two other criteria, namely the commercial availability and the financial cost of the individual elements of these clusters. We here develop a small representative library in which we to seek to satisfy the somewhat competing criteria of coverage (‘representativeness’), availability and cost. It is intended that the library chosen might serve as a testbed of molecules that may or may not be substrates for known or orphan drug transporters. A supplementary spreadsheet provides details, and their availability via a particular supplier.
Collapse
|
44
|
Billington S, Shoner S, Lee S, Clark-Snustad K, Pennington M, Lewis D, Muzi M, Rene S, Lee J, Nguyen TB, Kumar V, Ishida K, Chen L, Chu X, Lai Y, Salphati L, Hop CECA, Xiao G, Liao M, Unadkat JD. Positron Emission Tomography Imaging of [ 11 C]Rosuvastatin Hepatic Concentrations and Hepatobiliary Transport in Humans in the Absence and Presence of Cyclosporin A. Clin Pharmacol Ther 2019; 106:1056-1066. [PMID: 31102467 DOI: 10.1002/cpt.1506] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/25/2019] [Indexed: 01/16/2023]
Abstract
Using positron emission tomography imaging, we determined the hepatic concentrations and hepatobiliary transport of [11 C]rosuvastatin (RSV; i.v. injection) in the absence (n = 6) and presence (n = 4 of 6) of cyclosporin A (CsA; i.v. infusion) following a therapeutic dose of unlabeled RSV (5 mg, p.o.) in healthy human volunteers. The sinusoidal uptake, sinusoidal efflux, and biliary efflux clearance (CL; mL/minute) of [11 C]RSV, estimated through compartment modeling were 1,205.6 ± 384.8, 16.2 ± 11.2, and 5.1 ± 1.8, respectively (n = 6). CsA (blood concentration: 2.77 ± 0.24 μM), an organic-anion-transporting polypeptide, Na+ -taurocholate cotransporting polypeptide, and breast cancer resistance protein inhibitor increased [11 C]RSV systemic blood exposure (45%; P < 0.05), reduced its biliary efflux CL (52%; P < 0.05) and hepatic uptake (25%; P > 0.05) but did not affect its distribution into the kidneys. CsA increased plasma concentrations of coproporphyrin I and III and total bilirubin by 297 ± 69%, 384 ± 102%, and 81 ± 39%, respectively (P < 0.05). These data can be used in the future to verify predictions of hepatic concentrations and hepatobiliary transport of RSV.
Collapse
Affiliation(s)
- Sarah Billington
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.,Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals (Europe) Ltd., Abingdon-on-Thames, UK
| | - Steven Shoner
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Scott Lee
- Inflammatory Bowel Disease Program, University of Washington, Seattle, Washington, USA
| | - Kindra Clark-Snustad
- Inflammatory Bowel Disease Program, University of Washington, Seattle, Washington, USA
| | - Matthew Pennington
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, Washington, USA
| | - David Lewis
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Mark Muzi
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Shirley Rene
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Jean Lee
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Tot Bui Nguyen
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Vineet Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Kazuya Ishida
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.,Pharmacokinetics and Drug Metabolism, Amgen, Cambridge, Massachusetts, USA
| | - Laigao Chen
- Early Clinical Development, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts, USA
| | - Xiaoyan Chu
- Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey, USA
| | - Yurong Lai
- Department of Drug Metabolism, Gilead Sciences, Inc., Foster City, California, USA
| | - Laurent Salphati
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Cornelis E C A Hop
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Guangqing Xiao
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, Massachusetts, USA.,Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts, USA
| | - Mingxiang Liao
- Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
45
|
Jung KH, Oh SJ, Kang KJ, Han SJ, Nam KR, Park JA, Lee KC, Lee YJ, Choi JY. Effects of P-gp and Bcrp as brain efflux transporters on the uptake of [ 18 F]FPEB in the murine brain. Synapse 2019; 73:e22123. [PMID: 31269310 DOI: 10.1002/syn.22123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/11/2019] [Accepted: 06/28/2019] [Indexed: 01/16/2023]
Abstract
The purpose of this study was to determine whether the brain uptake of [18 F]FPEB is influenced by P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp) as efflux transporters in rodents. To assess this possible modulation, positron emission tomography studies were performed in animal models of pharmacological or genetic ablation of these transporters. Compared with the control conditions, when P-gp was blocked with tariquidar, there was an 8%-12% increase in the brain uptake of [18 F]FPEB. In P-gp knockout mice, such as Mdr1a/b(-/-) and Mdr1a/b(-/-) Bcrp1(-/-) , genetic ablation models, there was an increment of 8%-53% in [18 F]FPEB uptake compared with that in the wild-type mice. In contrast, Bcrp knockout mice showed a decrement of 5%-12% uptake and P-gp/Bcrp knockout group displayed an increment of 5%-17% compared with wild type. These results indicate that [18 F]FPEB is possibly a weak substrate for P-gp.
Collapse
Affiliation(s)
- Ki-Hye Jung
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sang Jin Han
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Ji Ae Park
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Bauer M, Tournier N, Langer O. Imaging P-Glycoprotein Function at the Blood-Brain Barrier as a Determinant of the Variability in Response to Central Nervous System Drugs. Clin Pharmacol Ther 2019; 105:1061-1064. [PMID: 30903694 PMCID: PMC6491962 DOI: 10.1002/cpt.1402] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/03/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nicolas Tournier
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, 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
|
49
|
Gessner A, König J, Fromm MF. Clinical Aspects of Transporter-Mediated Drug-Drug Interactions. Clin Pharmacol Ther 2019; 105:1386-1394. [PMID: 30648735 DOI: 10.1002/cpt.1360] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
Abstract
Drug transporters play an essential role in disposition and effects of multiple drugs. Plasma concentrations of the victim drug can be modified by drug-drug interactions occurring in enterocytes (e.g., P-glycoprotein), hepatocytes (e.g., organic anion-transporting polypeptide 1B1 (OATP1B1)), and/or renal proximal tubular cells (e.g., organic cation transporter 2 (OCT2)/multidrug and toxin extrusion 1 and 2-K (MATE1/MATE2-K)). In addition, transporter-mediated drug-drug interactions can cause altered local tissue concentrations and possibly altered effects/toxicity (e.g., in liver and kidneys). During drug development, there is now an intensive in vitro screening of new molecular entities as transporter substrates and inhibitors, followed if necessary by drug-drug interaction studies in healthy volunteers. Nevertheless, there are still unresolved issues, which will also be discussed in this review article (e.g., the clinical significance of transporter-mediated drug-drug interactions of particular relevance to the elderly who are prescribed multiple drugs, with additional impaired liver or kidney function, and the extent to which medication safety in real life could be improved by a reduction of those interactions).
Collapse
Affiliation(s)
- Arne Gessner
- 1Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jörg König
- 1Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F Fromm
- 1Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
50
|
Vodovar D, Aboab J, Silva S, Tournier N. Comprendre la tomographie par émission de positons (TEP) — une modalité d’imagerie pour la réanimation ? MEDECINE INTENSIVE REANIMATION 2019. [DOI: 10.3166/rea-2019-0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|