1
|
Salvi de Souza G, Mossel P, Somsen JF, Providência L, Bartels AL, Willemsen ATM, Dierckx RAJO, Furini CRG, Lammertsma AA, Tsoumpas C, Luurtsema G. Evaluating image-derived input functions for cerebral [ 18F]MC225 PET studies. FRONTIERS IN NUCLEAR MEDICINE 2025; 5:1597902. [PMID: 40538986 PMCID: PMC12176838 DOI: 10.3389/fnume.2025.1597902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 05/19/2025] [Indexed: 06/22/2025]
Abstract
Kinetic modelling of brain PET data is crucial for estimating quantitative biological parameters, traditionally requiring arterial sampling. This study evaluated whether arterial samples could be omitted to estimate the image-derived input function (IDIF) using a long axial field-of-view PET scanner. The use of internal carotid arteries (ICA) for IDIF estimation, along with venous samples for plasma-to-whole blood ratios and plasma parent fractions, was also assessed. Six healthy volunteers underwent [18F]MC225 scans with manual arterial sampling. IDIFs were derived from the aortic arch (IDIFAA) and calibrated using manual arterial samples (IDIFAA_CAL). ICA-derived IDIF was also calibrated (IDIFCA_CAL) and compared to IDIFAA_CAL. In a separate group of six volunteers, venous and arterial samples were collected to evaluate plasma-to-whole blood ratios, plasma parent fractions, and IDIF calibration (IDIFCA_CAL_VEN). Volume of distribution (VT) of different brain regions was estimated for all IDIFs techniques, corrected for plasma-to-whole blood ratio and plasma parent fraction (IDIFAA,P, IDIFAA_CAL,P, IDIFICA_CAL,P and IDIFICA_CAL_VEN_P). Our findings revealed discrepancies between IDIFAA and arterial samples, highlighting the importance of calibration. The differences between IDIFAA,P and IDIFAA_CAL,P were 9.2% for area under the curve and 4.0% for brain VT. IDIFICA_CAL,P showed strong agreement with IDIFA_CAL,P, with 1.2% VT difference. Venous sampling showed consistent agreement with arterial sampling for plasma parameters but was unreliable for IDIF calibration, leading to 39% VT differences. This study emphasises that arterial samples are still required for IDIF calibration and reliable VT estimation for [18F]MC225 PET tracer. ICA-derived IDIF, when calibrated, provides reliable VT estimates. Venous sampling is a potential alternative for estimating plasma parameters, but it is unsuitable for IDIF calibration. Trial Registry NCT05618119 (clinicaltrials.gov/study/NCT05618119).
Collapse
Affiliation(s)
- Giordana Salvi de Souza
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Pascalle Mossel
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Joost F. Somsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Laura Providência
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Anna L. Bartels
- Department of Neurology, Ommelander Ziekenhuis Groep, Scheemda, Netherlands
| | - Antoon T. M. Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Rudi A. J. O. Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Cristiane R. G. Furini
- School of Medicine, PUCRS, Porto Alegre, Brazil
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, PUCRS, Porto Alegre, Brazil
| | - Adriaan A. Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
2
|
Mossel P, Salvi de Souza G, Willemsen ATM, Stormezand GN, Colabufo NA, Toyohara J, Boersma HH, Dierckx RAJO, Lammertsma AA, Bartels AL, Luurtsema G. Measurement of cyclosporin induced changes in P-glycoprotein function at the human blood-brain barrier using [ 18F]MC225 and PET. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07320-0. [PMID: 40338304 DOI: 10.1007/s00259-025-07320-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION P-glycoprotein (P-gp) or multidrug-resistance protein is one of the most extensively studied efflux transporters at the blood-brain barrier (BBB). Changes in P-gp function are associated with several neurodegenerative and psychiatric diseases, including Alzheimer's disease, Parkinson's disease and schizophrenia and with the bioavailability of several pharmaceuticals in the brain, causing multi-drug resistance or side effects. PET imaging can be used to measure the P-gp function in vivo. This study aims to validate [18F]MC225 as specific P-gp PET tracer with the use of cyclosporin as selective P-gp inhibitor. METHODS Fourteen healthy volunteers (age 67 ± 5y) were included. Subjects underwent twice a 60 min dynamic [18F]MC225 (200MBq) PET scan with continuous arterial blood sampling and a cerebral T1-weighted MRI as anatomical reference. During the second scan, in five subjects, cyclosporin was administered in a dose of 2.5 mg/kg/hour, starting 30 min prior to the scan, to inhibit the BBB P-gp function. Tissue time-activity curves of preselected brain regions (Hammer's atlas) were fitted to a reversible two-tissue compartment model (2T4k) using the metabolite corrected plasma and uncorrected whole blood curves as input functions. RESULTS No significant difference was found in plasma kinetics, plasma curves, plasma-to-whole blood ratio, and the parent fraction of the baseline scans and scans after administration of cyclosporin. Volume of distribution values in whole brain grey matter showed a significant increase (6.18 ± 1.29 to 9.00 ± 1.29 mL·cm- 3,p = 0.03) after the administration of cyclosporin. CONCLUSION The outcomes of the current study reflect the potential ability of [18F]MC225 to measure cyclosporin induced changes in P-gp function at the human BBB in vivo. TRIAL REGISTRATION EudraCT 2020-001564-28.
Collapse
Affiliation(s)
- Pascalle Mossel
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Giordana Salvi de Souza
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Antoon T M Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gilles N Stormezand
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nicola A Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "A. Moro", via Orabona, 4, 70125, Bari, Italy
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 1730015, Japan
| | - Hendrikus H Boersma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna L Bartels
- Department of Neurology, Ommelander Ziekenhuis Groep, Scheemda, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
3
|
Tournier N, Langer O. Imaging the Activity of Efflux Transporters at the Blood-Brain Barrier in Neurologic Diseases: Radiotracer Selection Criteria. J Nucl Med 2025; 66:676-680. [PMID: 40015923 PMCID: PMC12051769 DOI: 10.2967/jnumed.124.269322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025] Open
Abstract
Efflux transporters of the adenosine triphosphate-binding cassette (ABC) superfamily, such as P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2), are highly expressed at the blood-brain barrier (BBB), where they contribute to maintaining brain homeostasis. P-gp may serve as an imaging biomarker to assess the contribution of BBB functionality rather than integrity to the onset or progression of various neurologic diseases. Considerable efforts have been made to develop radiolabeled P-gp substrates to assess cerebral P-gp activity with PET. However, initially developed radiotracers have limited clinical utility as they lack sensitivity to detect moderate, physiologically relevant changes in cerebral P-gp activity. Learning from this molecular imaging area has called for specific criteria, different from those classically used for other central nervous system targets, for developing and selecting suitable PET tracers to study ABC transporter activity at the BBB in different neurologic diseases.
Collapse
Affiliation(s)
- Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, INSERM, Orsay, France;
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; and
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Runwal P, Pyun J, Newman SA, Mawal C, Bush AI, Koehn LM, Nicolazzo JA. Ferric Ammonium Citrate Reduces Claudin-5 Abundance and Function in Primary Mouse Brain Endothelial Cells. Pharm Res 2025; 42:319-334. [PMID: 39939558 PMCID: PMC11880181 DOI: 10.1007/s11095-025-03826-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Iron overload is implicated in many neurodegenerative diseases, where there is also blood-brain barrier (BBB) dysfunction. As there is a growing interest in the role of iron in modulating key BBB proteins, this study assessed the effect of iron on the expression and function of P-glycoprotein (P-gp), breast cancer resistance protein (BCRP) and claudin-5 in primary mouse brain endothelial cells (MBECs) and their abundance in mouse brain microvessel-enriched membrane fractions (MVEFs). METHODS Following a 48 h treatment with ferric ammonium citrate (FAC, 250 µM), MBEC protein abundance (P-gp, BCRP and claudin-5) and mRNA (abcb1a, abcg2, and cldn5) were assessed by western blotting and RT-qPCR, respectively. Protein function was evaluated by assessing transport of substrates 3H-digoxin (P-gp), 3H-prazosin (BCRP) and 14C-sucrose (paracellular permeability). C57BL/6 mice received iron dextran (100 mg/kg, intraperitoneally) over 4 weeks, and MVEF protein abundance and iron levels (in MVEFs and plasma) were quantified via western blotting and inductively coupled plasma-mass spectrometry (ICP-MS), respectively. RESULTS FAC treatment reduced P-gp protein by 50% and abcb1a mRNA by 43%, without affecting 3H-digoxin transport. FAC did not alter BCRP protein or function, but decreased abcg2 mRNA by 59%. FAC reduced claudin-5 protein and cldn5 mRNA by 65% and 70%, respectively, resulting in a 200% increase in 14C-sucrose permeability. In vivo, iron dextran treatment significantly elevated plasma iron levels (2.2-fold) but did not affect brain MVEF iron content or alter P-gp, BCRP or claudin-5 protein abundance. CONCLUSIONS Iron overload modulates BBB transporters and junction proteins in vitro, highlighting potential implications for CNS drug delivery in neurodegenerative diseases.
Collapse
MESH Headings
- Animals
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Claudin-5/metabolism
- Claudin-5/genetics
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/drug effects
- Mice, Inbred C57BL
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- Brain/metabolism
- Brain/drug effects
- Brain/cytology
- Ferric Compounds/pharmacology
- Mice
- Quaternary Ammonium Compounds/pharmacology
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- Male
- Cells, Cultured
- Iron/metabolism
Collapse
Affiliation(s)
- Pranav Runwal
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jae Pyun
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Stephanie A Newman
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Celeste Mawal
- Oxidation Biology Lab, Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ashley I Bush
- Oxidation Biology Lab, Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- University of Melbourne, Parkville, VIC, Australia
| | - Liam M Koehn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
5
|
Salvi de Souza G, Furini CRG, Sijbesma JWA, Kominia M, Doorduin J, Giacobbo BL, Lammertsma AA, Tsoumpas C, Luurtsema G. Oral Administration of [ 18F]MC225 for Quantification of P-glycoprotein Function: A Feasibility Study. Mol Imaging Biol 2025; 27:89-98. [PMID: 39810067 PMCID: PMC11805767 DOI: 10.1007/s11307-024-01975-1] [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: 07/25/2024] [Revised: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE This preclinical study explored the feasibility of assessing P-glycoprotein (P-gp) function in both brain and gastrointestinal (GI) tract of rats using positron emission tomography (PET) following oral administration of [18F]MC225. Different oral administration protocols were evaluated, and radioactivity uptake was compared with uptake following intravenous administration. PROCEDURES Twelve male Wistar rats were divided into four groups and subjected to intravenous or oral [18F]MC225 administration protocols: G1 (intravenous route), G2 (oral administration without fasting), G3 (oral administration with fasting), and G4 (oral administration with fasting following administration of the P-gp inhibitor tariquidar). Dynamic brain imaging, late abdominal imaging, ex vivo biodistribution, and metabolite analysis were conducted to assess tracer distribution. RESULTS In the brain, oral administration yielded lower values compared with intravenous administration, resulting in a reduction in the tissue-to-plasma ratio by approximately 51% for the cortex and 45% for the midbrain and cerebellum. Fasting improved radioactivity uptake, aiding brain visualization. Unexpectedly, administration of the P-gp inhibitor tariquidar did not increase brain concentration, suggesting a signal that was dominated by non-specific uptake, possibly due to instability of [18F]MC225 in the GI tract. Metabolite analysis in G4 indicated a significant presence of polar metabolites. CONCLUSIONS Oral administration of [18F]MC225 faces challenges and, at this stage, cannot be used to quantify P-gp function. Further research to assess tracer stability and metabolism in the stomach and intestine will be essential for advancing the feasibility of oral tracer administration.
Collapse
Affiliation(s)
- Giordana Salvi de Souza
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- School of Medicine, PUCRS, Porto Alegre, Brazil
| | - Cristiane R G Furini
- School of Medicine, PUCRS, Porto Alegre, Brazil
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, PUCRS, Porto Alegre, Brazil
| | - Jürgen W A Sijbesma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maria Kominia
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bruno Lima Giacobbo
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
6
|
Liu Y, Ma J, Zhang Q, Wang Y, Sun Q. Mechanism of Metal Complexes in Alzheimer's Disease. Int J Mol Sci 2024; 25:11873. [PMID: 39595941 PMCID: PMC11593898 DOI: 10.3390/ijms252211873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a kind of neurodegenerative diseases characterized by beta-amyloid deposition and neurofibrillary tangles and is also the main cause of dementia. According to statistics, the incidence of AD is constantly increasing, bringing a great burden to individuals and society. Nonetheless, there is no cure for AD, and the available drugs are very limited apart from cholinesterase inhibitors and N-Methyl-D-aspartic acid (NMDA) antagonists, which merely alleviate symptoms without delaying the progression of the disease. Therefore, there is an urgent need to develop a medicine that can delay the progression of AD or cure it. In recent years, increasing evidence suggests that metal complexes have the enormous potential to treat AD through inhibiting the aggregation and cytotoxicity of Aβ, interfering with the congregation and hyperphosphorylation of tau, regulating dysfunctional synaptic and unbalanced neurotransmitters, etc. In this review, we summarize the current metal complexes and their mechanisms of action for treating AD, including ruthenium, platinum, zinc, vanadium, copper, magnesium, and other complexes.
Collapse
Affiliation(s)
- Yi Liu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; (Y.L.); (J.M.)
| | - Jiaying Ma
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; (Y.L.); (J.M.)
| | - Qianling Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518055, China;
| | - Yi Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518055, China;
| | - Qi Sun
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; (Y.L.); (J.M.)
| |
Collapse
|
7
|
Biali ME, Breuil L, Jackwerth M, Mairinger S, Weber M, Wölfl-Duchek M, Bamminger K, Rausch I, Nics L, Hacker M, Rodrigo S, Bouilleret V, Zeitlinger M, Pataraia E, Tournier N, Bauer M, Langer O. [ 11C]Metoclopramide PET can detect a seizure-induced up-regulation of cerebral P-glycoprotein in epilepsy patients. Fluids Barriers CNS 2024; 21:87. [PMID: 39465417 PMCID: PMC11514750 DOI: 10.1186/s12987-024-00588-8] [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: 08/19/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND P-glycoprotein (P-gp) is an efflux transporter which is abundantly expressed at the blood-brain barrier (BBB) and which has been implicated in the pathophysiology of various brain diseases. The radiolabelled antiemetic drug [11C]metoclopramide is a P-gp substrate for positron emission tomography (PET) imaging of P-gp function at the BBB. To assess whether [11C]metoclopramide can detect increased P-gp function in the human brain, we employed drug-resistant temporal lobe epilepsy (TLE) as a model disease with a well characterised, regional P-gp up-regulation at the BBB. METHODS Eight patients with drug-resistant (DRE) TLE, 5 seizure-free patients with drug-sensitive (DSE) focal epilepsy, and 15 healthy subjects underwent brain PET imaging with [11C]metoclopramide on a fully-integrated PET/MRI system. Concurrent with PET, arterial blood sampling was performed to generate a metabolite-corrected arterial plasma input function for kinetic modelling. The choroid plexus was outmasked on the PET images to remove signal contamination from the neighbouring hippocampus. Using a brain atlas, 10 temporal lobe sub-regions were defined and analysed with a 1-tissue-2-rate constant compartmental model to estimate the rate constants for radiotracer transfer from plasma to brain (K1) and from brain to plasma (k2), and the total volume of distribution (VT = K1/k2). RESULTS DRE patients but not DSE patients showed significantly higher k2 values and a trend towards lower VT values in several temporal lobe sub-regions located ipsilateral to the epileptic focus as compared to healthy subjects (k2: hippocampus: +34%, anterior temporal lobe, medial part: +28%, superior temporal gyrus, posterior part: +21%). CONCLUSIONS [11C]Metoclopramide PET can detect a seizure-induced P-gp up-regulation in the epileptic brain. The efflux rate constant k2 seems to be the most sensitive parameter to measure increased P-gp function with [11C]metoclopramide. Our study provides evidence that disease-induced alterations in P-gp expression at the BBB can lead to changes in the distribution of a central nervous system-active drug to the human brain, which could affect the efficacy and/or safety of drugs. [11C]Metoclopramide PET may be used to assess or predict the contribution of increased P-gp function to drug resistance and disease pathophysiology in various brain diseases. TRIAL REGISTRATION EudraCT 2019-003137-42. Registered 28 February 2020.
Collapse
Affiliation(s)
- Myriam El Biali
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Geneva, Switzerland
| | - Louise Breuil
- Laboratoire d'Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France
| | - 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 und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Wölfl-Duchek
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, 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
| | - Ivo Rausch
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Sebastian Rodrigo
- Laboratoire d'Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Viviane Bouilleret
- Laboratoire d'Imagerie Biomédicale Multimodale (BIOMAPS), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France
- Neurophysiologie et Epileptologie, Université Paris Saclay-APHP, Le Kremlin Bicêtre, Paris, France
| | - Markus Zeitlinger
- 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
| | - Martin Bauer
- 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 und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Kiełbowski K, Król M, Bakinowska E, Pawlik A. The Role of ABCB1, ABCG2, and SLC Transporters in Pharmacokinetic Parameters of Selected Drugs and Their Involvement in Drug-Drug Interactions. MEMBRANES 2024; 14:223. [PMID: 39590609 PMCID: PMC11596214 DOI: 10.3390/membranes14110223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024]
Abstract
Membrane transporters are expressed in a wide range of tissues in the human organism. These proteins regulate the penetration of various substances such as simple ions, xenobiotics, and an extensive number of therapeutics. ABC and SLC drug transporters play a crucial role in drug absorption, distribution, and elimination. Recent decades have shown their contribution to the systemic exposure and tissue penetration of numerous drugs, thereby having an impact on pharmacokinetic and pharmacodynamic parameters. Importantly, the activity and expression of these transporters depend on numerous conditions, including intestinal microbiome profiles or health conditions. Moreover, the combined intake of other drugs or natural agents further affects the functionality of these proteins. In this review, we will discuss the involvement of ABC and SLC transporters in drug disposition. Moreover, we will present current evidence of the potential role of drug transporters as therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (M.K.); (E.B.)
| |
Collapse
|
9
|
Liu W, Mossel P, Schwach V, Slart RHJA, Luurtsema G. Cardiac PET Imaging of ATP Binding Cassette (ABC) Transporters: Opportunities and Challenges. Pharmaceuticals (Basel) 2023; 16:1715. [PMID: 38139840 PMCID: PMC10748140 DOI: 10.3390/ph16121715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Adenosine triphosphate binding cassette (ABC) transporters are a broad family of membrane protein complexes that use energy to transport molecules across cells and/or intracellular organelle lipid membranes. Many drugs used to treat cardiac diseases have an affinity for these transporters. Among others, P-glycoprotein (P-gp) plays an essential role in regulating drug concentrations that reach cardiac tissue and therefore contribute to cardiotoxicity. As a molecular imaging modality, positron emission tomography (PET) has emerged as a viable technique to investigate the function of P-gp in organs and tissues. Using PET imaging to evaluate cardiac P-gp function provides new insights for drug development and improves the precise use of medications. Nevertheless, information in this field is limited. In this review, we aim to examine the current applications of ABC transporter PET imaging and its tracers in the heart, with a specific emphasis on P-gp. Furthermore, the opportunities and challenges in this novel field will be discussed.
Collapse
Affiliation(s)
- Wanling Liu
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
| | - Pascalle Mossel
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherlands;
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
- Department of Biomedical Photonic Imaging, University of Twente, 7500 AE Enschede, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (W.L.); (P.M.)
| |
Collapse
|