1
|
Lee SJ, Pearson TD, Dhaynaut M, MacDonagh AC, Wey HY, Wilks MQ, Roth BL, Hooker JM, Normandin MD. Selective Mu-Opioid Receptor Imaging Using 18F-Labeled Carfentanils. J Med Chem 2025; 68:1632-1644. [PMID: 39772615 DOI: 10.1021/acs.jmedchem.4c02287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Carfentanil, a highly potent synthetic opioid, paradoxically serves as a crucial positron emission tomography (PET) imaging tool in neurobiological studies of the mu-opioid receptor (MOR) system when labeled with carbon-11 ([11C]CFN). However, its clinical research use is hindered by extreme potency and the limited availability of short-lived carbon-11 (t1/2 = 20.4 min). We present fluorine-18-labeled fluorocarfentanils ([18F]FCFNs), which can be produced at higher molar activity, allowing for lower mass doses and benefiting from the longer half-life of fluorine-18 (t1/2 = 109.8 min), facilitating broader accessibility. Using copper-mediated radiofluorination, we synthesized a small [18F]FCFN library and conducted preclinical imaging evaluations. Two candidates, o-18F-1 and p-18F-2, showed optimal brain uptake, favorable pharmacokinetics, and high MOR-specific binding. Selectivity was confirmed through in vitro binding assays and in vivo PET scans. These [18F]FCFNs are promising for accessible human brain MOR imaging.
Collapse
Affiliation(s)
- So Jeong Lee
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Torben D Pearson
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Alexander C MacDonagh
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Moses Q Wilks
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
2
|
Soyer A, Leterrier S, Goutal S, Corvo C, Saba W, Caillé F, Bo GD, Winkeler A, Thibault K, Leroy C, Tournier N. Decreased opioid receptor availability and impaired neurometabolic coupling as signatures of morphine tolerance in male rats: A positron emission tomography study. Biomed Pharmacother 2025; 183:117848. [PMID: 39823723 DOI: 10.1016/j.biopha.2025.117848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/20/2025] Open
Abstract
Translational neuroimaging techniques are needed to address the impact of opioid tolerance on brain function and quantitatively monitor the impaired neuropharmacological response to opioids at the CNS level. A multiparametric PET study was conducted in rats. Rats received morphine daily to induce tolerance (15 mg/kg/day for 5 days), followed by 2-day withdrawal. Then, opioid effects were precipitated using a buprenorphine challenge (0.1 mg/kg, s.c, BUP-challenge), which safely enables full occupancy of available mu-opioid receptors (MOR). The impact of the BUP-challenge on the pain threshold was estimated using the hot-plate test. The corresponding availability of MOR was estimated using [11C]buprenorphine PET imaging (n = 4). The brain glucose metabolism was investigated using [18F]2-fluoro-D-deoxy-glucose ([18F]FDG) PET imaging after the BUP-challenge or saline (n = 5-6). Opioid tolerance was confirmed by the attenuated antinociceptive response to the BUP-challenge in morphine-treated rats compared to saline controls (p < 0.001). In tolerant rats, [11C]buprenorphine binding was decreased in MOR-rich regions (p < 0.01), and the baseline uptake of [18F]FDG was decreased (p < 0.05). The BUP-challenge decreased [18F]FDG uptake to a lower extent in tolerant rats compared with opioid-naive animals (p < 0.05), suggesting impaired neurometabolic coupling. Moreover, the impact of the BUP-challenge on the neurometabolic connectivity across brain regions was disrupted by opioid tolerance. PET imaging enables the study of the decreased availability of MOR and impaired neurometabolic coupling as molecular signatures of opioid tolerance in rats. Combining molecular neuroimaging with a suitable pharmacological challenge may provide a translational and quantitative paradigm to explore opioid tolerance at the CNS level in parallel to pharmacodynamics.
Collapse
Affiliation(s)
- Amélie Soyer
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sarah Leterrier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sébastien Goutal
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Cassandre Corvo
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Wadad Saba
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Fabien Caillé
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Gregory Dal Bo
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France; Department of Radiation Biological Effects, Armed Forces Biomedical Research Institute, Bretigny-sur-Orge 91220, France
| | - Alexandra Winkeler
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Karine Thibault
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France; Department of Radiation Biological Effects, Armed Forces Biomedical Research Institute, Bretigny-sur-Orge 91220, France
| | - Claire Leroy
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France.
| |
Collapse
|
3
|
Levinstein MR, Budinich RC, Bonaventura J, Schatzberg AF, Zarate CA, Michaelides M. Redefining Ketamine Pharmacology for Antidepressant Action: Synergistic NMDA and Opioid Receptor Interactions? Am J Psychiatry 2025:appiajp20240378. [PMID: 39810555 DOI: 10.1176/appi.ajp.20240378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Ketamine is a racemic compound and medication comprised of (S)-ketamine and (R)-ketamine enantiomers and its metabolites. It has been used for decades as a dissociative anesthetic, analgesic, and recreational drug. More recently, ketamine, its enantiomers, and its metabolites have been used or are being investigated for the treatment of refractory depression, as well as for comorbid disorders such as anxiety, obsessive-compulsive, and opioid use disorders. Despite its complex pharmacology, ketamine is referred to as an N-methyl-d-aspartate (NMDA) receptor antagonist. In this review, the authors argue that ketamine's pharmacology should be redefined to include opioid receptors and the endogenous opioid system. They also highlight a potential mechanism of action of ketamine for depression that is attributed to bifunctional, synergistic interactions involving NMDA and opioid receptors.
Collapse
Affiliation(s)
- Marjorie R Levinstein
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Section, NIDA, Baltimore (Levinstein, Budinich, Michaelides); Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford (Schatzberg); Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda (Zarate); Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (Michaelides)
| | - Reece C Budinich
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Section, NIDA, Baltimore (Levinstein, Budinich, Michaelides); Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford (Schatzberg); Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda (Zarate); Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (Michaelides)
| | - Jordi Bonaventura
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Section, NIDA, Baltimore (Levinstein, Budinich, Michaelides); Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford (Schatzberg); Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda (Zarate); Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (Michaelides)
| | - Alan F Schatzberg
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Section, NIDA, Baltimore (Levinstein, Budinich, Michaelides); Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford (Schatzberg); Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda (Zarate); Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (Michaelides)
| | - Carlos A Zarate
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Section, NIDA, Baltimore (Levinstein, Budinich, Michaelides); Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford (Schatzberg); Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda (Zarate); Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (Michaelides)
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Section, NIDA, Baltimore (Levinstein, Budinich, Michaelides); Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona (Bonaventura); Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford (Schatzberg); Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda (Zarate); Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (Michaelides)
| |
Collapse
|
4
|
Auvity S, Vodovar D, Goutal S, Cisternino S, Chevillard L, Soyer A, Bottlaender M, Caillé F, Mégarbane B, Tournier N. Brain PET imaging using 11C-flumazenil and 11C-buprenorphine does not support the hypothesis of a mutual interaction between buprenorphine and benzodiazepines at the neuroreceptor level. J Cereb Blood Flow Metab 2024; 44:449-458. [PMID: 38097513 PMCID: PMC10870960 DOI: 10.1177/0271678x231221040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 02/16/2024]
Abstract
Among opioids, buprenorphine presents a favorable safety profile with a limited risk of respiratory depression. However, fatalities have been reported when buprenorphine is combined to a benzodiazepine. Potentiation of buprenorphine interaction with opioid receptors (ORs) with benzodiazepines, and/or vice versa, is hypothesized to explain this drug-drug interaction (DDI). The mutual DDI between buprenorphine and benzodiazepines was investigated at the neuroreceptor level in nonhuman primates (n = 4 individuals) using brain PET imaging and kinetic modelling. The binding potential (BPND) of benzodiazepine receptor (BzR) was assessed using 11C-flumazenil PET imaging before and after administration of buprenorphine (0.2 mg, i.v.). Moreover, the brain kinetics and receptor binding of buprenorphine were investigated in the same individuals using 11C-buprenorphine PET imaging before and after administration of diazepam (10 mg, i.v.). Outcome parameters were compared using a two-way ANOVA. Buprenorphine did not impact the plasma nor brain kinetics of 11C-flumazenil. 11C-flumazenil BPND was unchanged following buprenorphine exposure, in any brain region (p > 0.05). Similarly, diazepam did not impact the plasma or brain kinetics of 11C-buprenorphine. 11C-buprenorphine volume of distribution (VT) was unchanged following diazepam exposure, in any brain region (p > 0.05). To conclude, our PET imaging findings do not support a neuropharmacokinetic or neuroreceptor-related mechanism of the buprenorphine/benzodiazepine interaction.
Collapse
Affiliation(s)
- Sylvain Auvity
- Faculté de Pharmacie, Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| | - Dominique Vodovar
- Faculté de Pharmacie, Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
- Réanimation Médicale et Toxicologique, Hôpital Lariboisière, Fédération de Toxicologie (APHP), 75010, Paris
| | - Sébastien Goutal
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Orsay, France
| | - Salvatore Cisternino
- Faculté de Pharmacie, Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| | - Lucie Chevillard
- Faculté de Pharmacie, Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| | - Amélie Soyer
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Orsay, France
| | - Fabien Caillé
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Orsay, France
| | - Bruno Mégarbane
- Faculté de Pharmacie, Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
- Réanimation Médicale et Toxicologique, Hôpital Lariboisière, Fédération de Toxicologie (APHP), 75010, Paris
| | - Nicolas Tournier
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Orsay, France
| |
Collapse
|
5
|
Leroy C, Goutal S, Breuil L, Gervais P, Cherkaoui H, Ciuciu P, Auvity S, Vodovar D, Comtat C, Lebon V, Bottlaender M, Tournier N. A pharmacological imaging challenge based on 11C-buprenorphine PET-MRI to explore the response to opioids in humans. Eur J Nucl Med Mol Imaging 2023; 50:3153-3154. [PMID: 37148295 DOI: 10.1007/s00259-023-06253-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Affiliation(s)
- Claire Leroy
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
| | - Sébastien Goutal
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
| | - Louise Breuil
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006, Paris, France
| | - Philippe Gervais
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
| | - Hamza Cherkaoui
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
- Parietal Team, Université Paris-Saclay, CEA, Inria, 91190, Gif-Sur-Yvette, France
| | - Philippe Ciuciu
- Parietal Team, Université Paris-Saclay, CEA, Inria, 91190, Gif-Sur-Yvette, France
| | - Sylvain Auvity
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006, Paris, France
| | - Dominique Vodovar
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006, Paris, France
| | - Claude Comtat
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
| | - Vincent Lebon
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France
- Université-Paris-Saclay, UNIACT, Neurospin, CEA, 91191, Gif-Sur-Yvette, France
| | - Nicolas Tournier
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401, Orsay, France.
| |
Collapse
|
6
|
Soyer A, Leterrier S, Breuil L, Goislard M, Leroy C, Saba W, Thibault K, Bo GD, Bottlaender M, Caillé F, Goutal S, Tournier N. Validation of a pharmacological imaging challenge using 11C-buprenorphine and 18F-2-fluoro-2-deoxy-D-glucose positron emission tomography to study the effects of buprenorphine to the rat brain. Front Neurosci 2023; 17:1181786. [PMID: 37234261 PMCID: PMC10205997 DOI: 10.3389/fnins.2023.1181786] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Aim Buprenorphine mainly acts as an agonist of mu-opioid receptors (mu-OR). High dose buprenorphine does not cause respiratory depression and can be safely administered to elicit typical opioid effects and explore pharmacodynamics. Acute buprenorphine, associated with functional and quantitative neuroimaging, may therefore provide a fully translational pharmacological challenge to explore the variability of response to opioids in vivo. We hypothesized that the CNS effects of acute buprenorphine could be monitored through changes in regional brain glucose metabolism, assessed using 18F-FDG microPET in rats. Materials and methods First, level of receptor occupancy associated with a single dose of buprenorphine (0.1 mg/kg, s.c) was investigated through blocking experiments using 11C-buprenorphine PET imaging. Behavioral study using the elevated plus-maze test (EPM) was performed to assess the impact of the selected dose on anxiety and also locomotor activity. Then, brain PET imaging using 18F-FDG was performed 30 min after injection of unlabeled buprenorphine (0.1 mg/kg, s.c) vs. saline. Two different 18F-FDG PET acquisition paradigms were compared: (i) 18F-FDG injected i.v. under anesthesia and (ii) 18F-FDG injected i.p. in awake animals to limit the impact of anesthesia. Results The selected dose of buprenorphine fully blocked the binding of 11C-buprenorphine in brain regions, suggesting complete receptor occupancy. This dose had no significant impact on behavioral tests used, regardless of the anesthetized/awake handling paradigm. In anesthetized rats, injection of unlabeled buprenorphine decreased the brain uptake of 18F-FDG in most brain regions except in the cerebellum which could be used as a normalization region. Buprenorphine treatment significantly decreased the normalized brain uptake of 18F-FDG in the thalamus, striatum and midbrain (p < 0.05), where binding of 11C-buprenorphine was the highest. The awake paradigm did not improve sensitivity and impact of buprenorphine on brain glucose metabolism could not be reliably estimated. Conclusion Buprenorphine (0.1 mg/kg, s.c) combined with 18F-FDG brain PET in isoflurane anesthetized rats provides a simple pharmacological imaging challenge to investigate the CNS effects of full receptor occupancy by this partial mu-OR agonist. Sensitivity of the method was not improved in awake animals. This strategy may be useful to investigate de desensitization of mu-OR associated with opioid tolerance in vivo.
Collapse
Affiliation(s)
- Amélie Soyer
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sarah Leterrier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Louise Breuil
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Maud Goislard
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Claire Leroy
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Wadad Saba
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Karine Thibault
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
- Department of Toxicology and Chemical Risks, Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Gregory Dal Bo
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
- Department of Toxicology and Chemical Risks, Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Michel Bottlaender
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Fabien Caillé
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sébastien Goutal
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| |
Collapse
|
7
|
Pawar SD, Gawali K, Kulhari H, Murty US, Kumar P. Amoxapine-Loaded Solid Lipid Nanoparticles with Superior Preclinical Pharmacokinetics for Better Brain Delivery: LC-MS/MS and GC-MS Analysis. ACS Chem Neurosci 2023. [PMID: 37027804 DOI: 10.1021/acschemneuro.2c00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023] Open
Abstract
The tricyclic antidepressant amoxapine (AMX) has been reported for a rapid onset of action compared to other cyclic antidepressants. It has very low solubility and bioavailability due to first-pass metabolism. Therefore, we planned to develop solid lipid nanoparticles (SLNs) of AMX using a single emulsification method to increase its solubility and bioavailability. HPLC and LC-MS/MS methods were developed further to quantify AMX in the formulation, plasma, and brain tissue samples. The formulation was studied for entrapment efficiency, loading, and in vitro drug release. Particle size and ζ potential analyses, AFM, SEM, TEM, DSC, and XRD were used for further characterization. In vivo oral pharmacokinetic and brain pharmacokinetic studies were performed using Wistar rats. The entrapment and loading efficiencies of AMX in SLNs were 85.8 ± 3.42 and 4.5 ± 0.45%, respectively. The developed formulation had a mean particle size of 151.5 ± 7.02 nm and a polydispersity index of 0.40 ± 0.11. DSC and XRD results indicated that AMX was incorporated into the nanocarrier system in an amorphous form. SEM, TEM, and AFM studies of AMX-SLNs confirmed the particles' spherical shape and nanoscale size. AMX solubility increased by approx. 2.67 times compared to the pure drug. The developed LC-MS/MS method was successfully applied to the oral and brain pharmacokinetic study of AMX-loaded SLNs in rats. Oral bioavailability was enhanced 1.6 times compared to the pure drug. The peak plasma concentrations of pure AMX and AMX-SLNs were 617.4 ± 137.4 and 1043.5 ± 150.2 (ng/mL), respectively. AMX-SLNs showed more than 5.8 times brain concentration compared to the pure drug. Based on the findings, it appears that utilizing a solid lipid nanoparticle carrier to transport AMX can be a highly effective delivery method with improved pharmacokinetic properties in the brain. This approach may prove valuable for future antidepressant treatment.
Collapse
Affiliation(s)
- Sachin Dattram Pawar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Post Office Changsari, Kamrup, Assam 781101, India
| | - Komal Gawali
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Post Office Changsari, Kamrup, Assam 781101, India
| | - Hitesh Kulhari
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Post Office Changsari, Kamrup, Assam 781101, India
- School of Nano Sciences, Central University of Gujarat, Gandhinagar 382030, India
| | - Upadhyayula Suryanarayana Murty
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Post Office Changsari, Kamrup, Assam 781101, India
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Post Office Changsari, Kamrup, Assam 781101, India
| |
Collapse
|
8
|
Bodnar RJ. Endogenous opiates and behavior: 2021. Peptides 2023; 164:171004. [PMID: 36990387 DOI: 10.1016/j.peptides.2023.171004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
This paper is the forty-fourth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2021 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonizts and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY 11367, USA.
| |
Collapse
|
9
|
Amirali AS, Hecker JC, Figueroa HM, Effinger DP, Montoro RA, Jedema HP, Vogt CD, Newman AH, Schindler CW, Bradberry CW. Effects of buprenorphine, methadone, and cariprazine on economic choice between remifentanil and food in squirrel monkeys. ADDICTION NEUROSCIENCE 2023; 5:100065. [PMID: 36873095 PMCID: PMC9979865 DOI: 10.1016/j.addicn.2023.100065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
We recently reported an economic choice task in which squirrel monkeys chose between differing amounts of remifentanil, a fast-acting opioid, or a food reward to develop a preclinical screen for evaluating potential pharmacotherapies for opioid dependence. Herein, two known opioid addiction treatments are evaluated using this task, as well as a potential new agent, cariprazine, a dopamine D2/D3 receptor partial agonist currently used to treat bipolar disorder and schizophrenia. Preclinical rodent studies suggest this class of compounds may reduce opiate self-administration. Squirrel monkeys were pretreated daily with clinically relevant doses of each compound during the five days of treatment evaluation using the economic choice task. Shifts in drug preference were measured as changes in subjects' indifference values, where the probability of drug and milk choice are equivalent. Buprenorphine produced a significant shift in indifference value between baseline and treatment weeks, indicating a decrease in drug preference. Subjects treated with methadone and cariprazine did not show any significant shift in drug preference. Differences between the buprenorphine and methadone results likely reflect a lack of opioid dependence in the subjects. The cariprazine results suggest that it does not alter opioid reward in non-dependent primates over a five day period.
Collapse
Affiliation(s)
- Alishan S. Amirali
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| | - Jacquelin C. Hecker
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| | - Hector M. Figueroa
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| | - Devin P. Effinger
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
- Department of Pharmacology, University of North Carolina Chapel Hill, Chapel Hill, NC
| | - Rodrigo A. Montoro
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
- Medical Scientist Training Program (MTSP) at UW-Madison, Madison, WI
| | - Hank P. Jedema
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| | - Caleb D. Vogt
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| | - Charles W. Schindler
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| | - Charles W. Bradberry
- Behavioral Neuroscience Branch, National Institute on Drug Abuse, Intramural research Program, Baltimore, MD, USA
| |
Collapse
|
10
|
Hakomäki H, Eskola S, Kokki H, Lehtonen M, Räsänen J, Laaksonen S, Voipio HM, Ranta VP, Kokki M. Central Nervous System Distribution of Buprenorphine in Pregnant Sheep, Fetuses and Newborn Lambs After Continuous Transdermal and Single Subcutaneous Extended-Release Dosing. Eur J Pharm Sci 2022; 178:106283. [PMID: 36029997 DOI: 10.1016/j.ejps.2022.106283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022]
Abstract
Buprenorphine is used during pregnancy for the treatment of opioid use disorder. Limited data exist on the central nervous system (CNS) permeation and distribution, and on the fetal exposure to buprenorphine. The aim of our study was to determine the extent of buprenorphine distribution to CNS in the pregnant sheep, and their fetus at steady-state, and their newborn lambs postdelivery, using three different dosing regimens. Twenty-eight pregnant ewes in late gestation received buprenorphine via 7-day transdermal patch releasing buprenorphine 20 µg/h (n=9) or 40 µg/h (n=11), or an extended-release 8 mg/week subcutaneous injection (n=8). Plasma, cerebrospinal fluid, and CNS tissue samples were collected at steady-state from ewes and fetuses, and from lambs 0.33 - 45 hours after delivery. High accumulation of buprenorphine was observed in all CNS tissues. The median CNS/plasma concentration -ratios of buprenorphine in different CNS areas ranged between 13 and 50 in the ewes, and between 26 and 198 in the fetuses. In the ewes the CNS/plasma -ratios were similar after the three dosing regimens, but higher in the fetuses in the 40 µg/h dosing group, medians 65 - 122, than in the 20 µg/h group, medians 26 - 54. The subcutaneous injection (theoretical release rate 47.6 µg/h) produced higher concentrations than observed after 40 µg/h transdermal patch dosing. The median fetal/maternal concentration -ratios in different dosing groups ranged between 0.21 and 0.54 in plasma, and between 0.38 and 1.3 in CNS tissues, respectively, with the highest ratios observed in the spinal cord. Buprenorphine concentrations in the cerebrospinal fluid were 8 - 13 % of the concurrent plasma concentration in the ewes and 28 % in the fetuses. Buprenorphine was quantifiable in the newborn lambs' plasma and CNS tissues two days postdelivery. Norbuprenorphine was analyzed from all plasma, cerebrospinal fluid, and CNS tissue samples but was nondetectable or below the LLOQ in most. The current study demonstrates that buprenorphine accumulates into CNS tissues at much higher concentrations than in plasma in pregnant sheep, fetuses, and their newborn lambs even 45 hours after delivery.
Collapse
Key Words
- BUP, Buprenorphine
- CL, Plasma clearance
- CNS, Central nervous system
- CSF, Cerebrospinal fluid
- F/M -ratio, Fetal to maternal concentration ratio
- HPLC, , High-performance liquid chromatography
- L/M -ratio, Lamb to maternal concentration ratio
- LC/MS/MS, Liquid chromatography - tandem mass spectrometry
- LLOQ, Lower limit of quantification
- NBUP, Norbuprenorphine
- brain
- buprenorphine
- pharmacokinetics, pregnancy
- sheep
- tissue
Collapse
Affiliation(s)
| | - Sophia Eskola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Hannu Kokki
- School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Juha Räsänen
- Fetal Medicine Center, Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sakari Laaksonen
- Department of Comparative Medicine, Oulu Laboratory Animal Centre, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Hanna-Marja Voipio
- Department of Comparative Medicine, Oulu Laboratory Animal Centre, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Veli-Pekka Ranta
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Merja Kokki
- Department of Anesthesiology and Intensive Care, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
11
|
Vermeulen I, Isin EM, Barton P, Cillero-Pastor B, Heeren RM. Multimodal molecular imaging in drug discovery and development. Drug Discov Today 2022; 27:2086-2099. [DOI: 10.1016/j.drudis.2022.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/03/2022] [Accepted: 04/08/2022] [Indexed: 02/06/2023]
|
12
|
Vodovar D, Chevillard L, Caillé F, Risède P, Pottier G, Auvity S, Mégarbane B, Tournier N. Mechanisms of respiratory depression induced by the combination of buprenorphine and diazepam in rats. Br J Anaesth 2022; 128:584-595. [PMID: 34872716 DOI: 10.1016/j.bja.2021.10.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/05/2021] [Accepted: 10/28/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The safety profile of buprenorphine has encouraged its widespread use. However, fatalities have been attributed to benzodiazepine/buprenorphine combinations, by poorly understood mechanisms of toxicity. Mechanistic hypotheses include (i) benzodiazepine-mediated increase in brain buprenorphine (pharmacokinetic hypothesis); (ii) benzodiazepine-mediated potentiation of buprenorphine interaction with opioid receptors (receptor hypothesis); and (iii) combined effects of buprenorphine and benzodiazepine on respiratory parameters (pharmacodynamic hypothesis). METHODS We studied the neuro-respiratory effects of buprenorphine (30 mg kg-1, i.p.), diazepam (20 mg kg-1, s.c.), and diazepam/buprenorphine combination in rats using arterial blood gas analysis, plethysmography, and diaphragm electromyography. Pretreatments with various opioid and gamma-aminobutyric acid receptor antagonists were tested. Diazepam impact on brain 11C-buprenorphine kinetics and binding to opioid receptors was studied using positron emission tomography imaging. RESULTS In contrast to diazepam and buprenorphine alone, diazepam/buprenorphine induced early-onset sedation (P<0.05) and respiratory depression (P<0.001). Diazepam did not alter 11C-buprenorphine brain kinetics or binding to opioid receptors. Diazepam/buprenorphine-induced effects on inspiratory time were additive, driven by buprenorphine (P<0.0001) and were blocked by naloxonazine (P<0.01). Diazepam/buprenorphine-induced effects on expiratory time were non-additive (P<0.001), different from buprenorphine-induced effects (P<0.05) and were blocked by flumazenil (P<0.01). Diazepam/buprenorphine-induced effects on tidal volume were non-additive (P<0.01), different from diazepam- (P<0.05) and buprenorphine-induced effects (P<0.0001) and were blocked by naloxonazine (P<0.05) and flumazenil (P<0.05). Compared with buprenorphine, diazepam/buprenorphine decreased diaphragm contraction amplitude (P<0.01). CONCLUSIONS Pharmacodynamic parameters and antagonist pretreatments indicate that diazepam/buprenorphine-induced respiratory depression results from a pharmacodynamic interaction between both drugs on ventilatory parameters.
Collapse
Affiliation(s)
- Dominique Vodovar
- Inserm UMRS-1144, Paris, France; Université de Paris, Paris, France; Université Paris-Saclay - CEA - CNRS - Inserm - BioMaps, Orsay, France; Paris Poison Center, Assistance Publique - Hôpitaux de Paris, Paris, France; Department of Medical and Toxicological Critical Care, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Lucie Chevillard
- Inserm UMRS-1144, Paris, France; Université de Paris, Paris, France
| | - Fabien Caillé
- Université Paris-Saclay - CEA - CNRS - Inserm - BioMaps, Orsay, France
| | | | - Géraldine Pottier
- Université Paris-Saclay - CEA - CNRS - Inserm - BioMaps, Orsay, France
| | - Sylvain Auvity
- Université Paris-Saclay - CEA - CNRS - Inserm - BioMaps, Orsay, France
| | - Bruno Mégarbane
- Inserm UMRS-1144, Paris, France; Université de Paris, Paris, France; Department of Medical and Toxicological Critical Care, Assistance Publique - Hôpitaux de Paris, Paris, France.
| | - Nicolas Tournier
- Université Paris-Saclay - CEA - CNRS - Inserm - BioMaps, Orsay, France
| |
Collapse
|