1
|
Mercier J, Bani M, Colson AO, Germani M, Lalla M, Plisson C, Huiban M, Searle G, Mathy FX, Nicholl R, Otoul C, Smit JW, van Asch V, Wagneur M, Maguire RP. Evaluation and Application of a PET Tracer in Preclinical and Phase 1 Studies to Determine the Brain Biodistribution of Minzasolmin (UCB0599). Mol Imaging Biol 2024; 26:310-321. [PMID: 38110790 DOI: 10.1007/s11307-023-01878-7] [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: 04/06/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023]
Abstract
PURPOSE Minzasolmin (UCB0599) is an orally administered, small molecule inhibitor of ASYN misfolding in development as a potential disease-modifying therapy for Parkinson's disease. Here we describe the preclinical development of a radiolabeled tracer and results from a phase 1 study using the tracer to investigate the brain distribution of minzasolmin. PROCEDURES In the preclinical study, two radiolabeling positions were investigated on the S-enantiomer of minzasolmin (UCB2713): [11C]methylamine UCB2713 ([11C-N-CH3]UCB2713) and [11C]carbonyl UCB2713 ([11C-CO]UCB2713). Male C57 black 6 mice (N = 10) received intravenous [11C-N-CH3]UCB2713; brain homogenates were assessed for radioactivity and plasma samples analyzed by high-performance liquid chromatography. Positron emission tomography-computed tomography (PET-CT) was used to image brains in a subset of mice (n = 3). In the open-label, phase 1 study, healthy volunteers were scanned twice with PET-CT following injection with [11C]minzasolmin radiotracer (≤ 10 µg), first without, then with oral dosing with non-radiolabeled minzasolmin 360 mg. PRIMARY OBJECTIVE to determine biodistribution of minzasolmin in the human brain; secondary objectives included minzasolmin safety/tolerability. RESULTS Preclinical data supported the use of [11C]minzasolmin in clinical studies. In the phase 1 study, PET data showed substantial drug signal in the brain of healthy volunteers (N = 4). The mean estimated whole brain total distribution volume (VT) at equilibrium across all regions of interest was 0.512 mL/cm3, no difference in VT was observed following administration of minzasolmin 360 mg. Treatment-emergent adverse events (TEAEs) were reported by 75% (n = 3) of participants. No drug-related TEAEs, deaths, serious adverse events, or discontinuations were reported. CONCLUSION Following positive preclinical results with the N-methyl labeled PET tracer, [11C]minzasolmin was used in the phase 1 study, which demonstrated that minzasolmin readily crossed the blood-brain barrier and was well distributed throughout the brain. Safety and pharmacokinetic findings were consistent with previous early-phase studies (such as UP0077, NCT04875962).
Collapse
Affiliation(s)
| | | | | | | | - Marianna Lalla
- UCB Pharma, Braine L'Alleud, Belgium
- OxSonics, Oxford, UK
| | | | | | | | | | | | | | - Johan Willem Smit
- UCB Pharma, Braine L'Alleud, Belgium
- Curare Consulting, Hamburg, Germany
| | | | | | | |
Collapse
|
2
|
Frautschi PC, Singh AP, Stowe NA, Yu JPJ. Multimodal Neuroimaging of the Effect of Serotonergic Psychedelics on the Brain. AJNR Am J Neuroradiol 2024; 45:ajnr.A8118. [PMID: 38360790 DOI: 10.3174/ajnr.a8118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/20/2023] [Indexed: 02/17/2024]
Abstract
The neurobiological mechanisms underpinning psychiatric disorders such as treatment-resistant major depression, post-traumatic stress disorder, and substance use disorders, remain unknown. Psychedelic compounds, such as psilocybin, lysergic acid diethylamide, and N,N-dimethyltryptamine, have emerged as potential therapies for these disorders because of their hypothesized ability to induce neuroplastic effects and alter functional networks in the brain. Yet, the mechanisms underpinning the neurobiological treatment response remain obscure. Quantitative neuroimaging is uniquely positioned to provide insight into the neurobiological mechanisms of these emerging therapies and quantify the patient treatment response. This review aims to synthesize our current state-of-the-art understanding of the functional changes occurring in the brain following psilocybin, lysergic acid diethylamide, or N,N-dimethyltryptamine administration in human participants with fMRI and PET. We further aim to disseminate our understanding of psychedelic compounds as they relate to neuroimaging with the goal of improved diagnostics and treatment of neuropsychiatric illness.
Collapse
Affiliation(s)
- Paloma C Frautschi
- Department of Radiology (P.C.F., A.P.S., J.-P.J.Y.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ajay P Singh
- Department of Radiology (P.C.F., A.P.S., J.-P.J.Y.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Graduate Program in Cellular and Molecular Biology (A.P.S., J.-P.J.Y.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Nicholas A Stowe
- Neuroscience Training Program, Wisconsin Institutes for Medical Research (N.A.S., J.-P.J.Y.), University of Wisconsin-Madison, Madison, Wisconsin
| | - John-Paul J Yu
- Department of Radiology (P.C.F., A.P.S., J.-P.J.Y.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Neuroscience Training Program, Wisconsin Institutes for Medical Research (N.A.S., J.-P.J.Y.), University of Wisconsin-Madison, Madison, Wisconsin
- Graduate Program in Cellular and Molecular Biology (A.P.S., J.-P.J.Y.), University of Wisconsin-Madison, Madison, Wisconsin
- Department of Biomedical Engineering (J.-P.J.Y.), University of Wisconsin-Madison, Madison, Wisconsin
- Department of Psychiatry (J.-P.J.Y.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
3
|
Shegani A, Kealey S, Luzi F, Basagni F, Machado JDM, Ekici SD, Ferocino A, Gee AD, Bongarzone S. Radiosynthesis, Preclinical, and Clinical Positron Emission Tomography Studies of Carbon-11 Labeled Endogenous and Natural Exogenous Compounds. Chem Rev 2023; 123:105-229. [PMID: 36399832 PMCID: PMC9837829 DOI: 10.1021/acs.chemrev.2c00398] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Indexed: 11/19/2022]
Abstract
The presence of positron emission tomography (PET) centers at most major hospitals worldwide, along with the improvement of PET scanner sensitivity and the introduction of total body PET systems, has increased the interest in the PET tracer development using the short-lived radionuclides carbon-11. In the last few decades, methodological improvements and fully automated modules have allowed the development of carbon-11 tracers for clinical use. Radiolabeling natural compounds with carbon-11 by substituting one of the backbone carbons with the radionuclide has provided important information on the biochemistry of the authentic compounds and increased the understanding of their in vivo behavior in healthy and diseased states. The number of endogenous and natural compounds essential for human life is staggering, ranging from simple alcohols to vitamins and peptides. This review collates all the carbon-11 radiolabeled endogenous and natural exogenous compounds synthesised to date, including essential information on their radiochemistry methodologies and preclinical and clinical studies in healthy subjects.
Collapse
Affiliation(s)
- Antonio Shegani
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Steven Kealey
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Federico Luzi
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Filippo Basagni
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum−University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Joana do Mar Machado
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Sevban Doğan Ekici
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Alessandra Ferocino
- Institute
of Organic Synthesis and Photoreactivity, Italian National Research Council, via Piero Gobetti 101, 40129 Bologna, Italy
| | - Antony D. Gee
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Salvatore Bongarzone
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
4
|
Fu H, Rong J, Chen Z, Zhou J, Collier T, Liang SH. Positron Emission Tomography (PET) Imaging Tracers for Serotonin Receptors. J Med Chem 2022; 65:10755-10808. [PMID: 35939391 DOI: 10.1021/acs.jmedchem.2c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) and 5-HT receptors (5-HTRs) have crucial roles in various neuropsychiatric disorders and neurodegenerative diseases, making them attractive diagnostic and therapeutic targets. Positron emission tomography (PET) is a noninvasive nuclear molecular imaging technique and is an essential tool in clinical diagnosis and drug discovery. In this context, numerous PET ligands have been developed for "visualizing" 5-HTRs in the brain and translated into human use to study disease mechanisms and/or support drug development. Herein, we present a comprehensive repertoire of 5-HTR PET ligands by focusing on their chemotypes and performance in PET imaging studies. Furthermore, this Perspective summarizes recent 5-HTR-focused drug discovery, including biased agonists and allosteric modulators, which would stimulate the development of more potent and subtype-selective 5-HTR PET ligands and thus further our understanding of 5-HTR biology.
Collapse
Affiliation(s)
- Hualong Fu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Zhen Chen
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Jingyin Zhou
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Thomas Collier
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
5
|
Mangeant R, Dubost E, Cailly T, Collot V. Radiotracers for the Central Serotoninergic System. Pharmaceuticals (Basel) 2022; 15:571. [PMID: 35631397 PMCID: PMC9143978 DOI: 10.3390/ph15050571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
This review lists the most important radiotracers described so far for imaging the central serotoninergic system. Single-photon emission computed tomography and positron emission tomography radiotracers are reviewed and critically discussed for each receptor.
Collapse
Affiliation(s)
- Reynald Mangeant
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| | - Emmanuelle Dubost
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| | - Thomas Cailly
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
- UNICAEN, IMOGERE, Normandie Univ., 14000 Caen, France
- CHU Côte de Nacre, Department of Nuclear Medicine, 14000 Caen, France
| | - Valérie Collot
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| |
Collapse
|
6
|
Märcher Rørsted E, Jensen AA, Kristensen JL. 25CN-NBOH: A Selective Agonist for in vitro and in vivo Investigations of the Serotonin 2A Receptor. ChemMedChem 2021; 16:3263-3270. [PMID: 34288515 DOI: 10.1002/cmdc.202100395] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Indexed: 01/20/2023]
Abstract
4-(2-((2-hydroxybenzyl)amino)ethyl)-2,5-dimethoxybenzonitrile (25CN-NBOH) was first reported as a potent and selective serotonin 2A receptor (5-HT2A R) agonist in 2014, and it has since found extensive use as a pharmacological tool in a variety of in vitro, ex vivo and in vivo studies. 25CN-NBOH is readily available from a synthetic perspective using standard chemical transformations, and displays favorable physiochemical properties in terms of stability and solubility. Due to its superior selectivity for 5-HT2A R, 25CN-NBOH has been used to investigate the effects of selective 5-HT2A R activation in vivo, and has thus become an important pharmacological tool for the exploration of 5-HT2A R signaling in a range of animal models. In the present review, we outline the discovery of 25CN-NBOH, its pharmacological profile and major findings from studies where it has been used.
Collapse
Affiliation(s)
- Emil Märcher Rørsted
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark
| | - Jesper L Kristensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark
| |
Collapse
|
7
|
Cumming P, Scheidegger M, Dornbierer D, Palner M, Quednow BB, Martin-Soelch C. Molecular and Functional Imaging Studies of Psychedelic Drug Action in Animals and Humans. Molecules 2021; 26:2451. [PMID: 33922330 PMCID: PMC8122807 DOI: 10.3390/molecules26092451] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
Hallucinogens are a loosely defined group of compounds including LSD, N,N-dimethyltryptamines, mescaline, psilocybin/psilocin, and 2,5-dimethoxy-4-methamphetamine (DOM), which can evoke intense visual and emotional experiences. We are witnessing a renaissance of research interest in hallucinogens, driven by increasing awareness of their psychotherapeutic potential. As such, we now present a narrative review of the literature on hallucinogen binding in vitro and ex vivo, and the various molecular imaging studies with positron emission tomography (PET) or single photon emission computer tomography (SPECT). In general, molecular imaging can depict the uptake and binding distribution of labelled hallucinogenic compounds or their congeners in the brain, as was shown in an early PET study with N1-([11C]-methyl)-2-bromo-LSD ([11C]-MBL); displacement with the non-radioactive competitor ketanserin confirmed that the majority of [11C]-MBL specific binding was to serotonin 5-HT2A receptors. However, interactions at serotonin 5HT1A and other classes of receptors and pleotropic effects on second messenger pathways may contribute to the particular experiential phenomenologies of LSD and other hallucinogenic compounds. Other salient aspects of hallucinogen action include permeability to the blood-brain barrier, the rates of metabolism and elimination, and the formation of active metabolites. Despite the maturation of radiochemistry and molecular imaging in recent years, there has been only a handful of PET or SPECT studies of radiolabeled hallucinogens, most recently using the 5-HT2A/2C agonist N-(2[11CH3O]-methoxybenzyl)-2,5-dimethoxy- 4-bromophenethylamine ([11C]Cimbi-36). In addition to PET studies of target engagement at neuroreceptors and transporters, there is a small number of studies on the effects of hallucinogenic compounds on cerebral perfusion ([15O]-water) or metabolism ([18F]-fluorodeoxyglucose/FDG). There remains considerable scope for basic imaging research on the sites of interaction of hallucinogens and their cerebrometabolic effects; we expect that hybrid imaging with PET in conjunction with functional magnetic resonance imaging (fMRI) should provide especially useful for the next phase of this research.
Collapse
Affiliation(s)
- Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane 4059, Australia
| | - Milan Scheidegger
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital of the University of Zurich, CH-8032 Zurich, Switzerland; (M.S.); (D.D.); (B.B.Q.)
| | - Dario Dornbierer
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital of the University of Zurich, CH-8032 Zurich, Switzerland; (M.S.); (D.D.); (B.B.Q.)
| | - Mikael Palner
- Odense Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark;
- Department of Nuclear Medicine, Odense University Hospital, DK-5000 Odense, Denmark
- Neurobiology Research Unit, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark
| | - Boris B. Quednow
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital of the University of Zurich, CH-8032 Zurich, Switzerland; (M.S.); (D.D.); (B.B.Q.)
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, CH-8058 Zurich, Switzerland
| | | |
Collapse
|
8
|
Zhang C, Yan Z, Maknojia A, Riquelme MA, Gu S, Booher G, Wallace DJ, Bartanusz V, Goswami A, Xiong W, Zhang N, Mader MJ, An Z, Sayre NL, Jiang JX. Inhibition of astrocyte hemichannel improves recovery from spinal cord injury. JCI Insight 2021; 6:134611. [PMID: 33682795 PMCID: PMC8021110 DOI: 10.1172/jci.insight.134611] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/27/2021] [Indexed: 11/25/2022] Open
Abstract
Spinal cord injury (SCI) causes severe disability, and the current inability to restore function to the damaged spinal cord leads to lasting detrimental consequences to patients. One strategy to reduce SCI morbidity involves limiting the spread of secondary damage after injury. Previous studies have shown that connexin 43 (Cx43), a gap junction protein richly expressed in spinal cord astrocytes, is a potential mediator of secondary damage. Here, we developed a specific inhibitory antibody, mouse-human chimeric MHC1 antibody (MHC1), that inhibited Cx43 hemichannels, but not gap junctions, and reduced secondary damage in 2 incomplete SCI mouse models. MHC1 inhibited the activation of Cx43 hemichannels in both primary spinal astrocytes and astrocytes in situ. In both SCI mouse models, administration of MHC1 after SCI significantly improved hind limb locomotion function. Remarkably, a single administration of MHC1 30 minutes after injury improved the recovery up to 8 weeks post-SCI. Moreover, MHC1 treatment decreased gliosis and lesion sizes, increased white and gray matter sparing, and improved neuronal survival. Together, these results suggest that inhibition of Cx43 hemichannel function after traumatic SCI reduces secondary damage, limits perilesional gliosis, and improves functional recovery. By targeting hemichannels specifically with an antibody, this study provides a potentially new, innovative therapeutic approach in treating SCI.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhao Yan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Asif Maknojia
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Grant Booher
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David J Wallace
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Viktor Bartanusz
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Akshay Goswami
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael J Mader
- Audie L. Murphy VA Hospital, South Texas Veteran's Health Care System, San Antonio, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Naomi L Sayre
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Audie L. Murphy VA Hospital, South Texas Veteran's Health Care System, San Antonio, Texas, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
9
|
Avedissian SN, Dyavar SR, Fox HS, Fletcher CV. Pharmacologic approaches to HIV-associated neurocognitive disorders. Curr Opin Pharmacol 2020; 54:102-108. [PMID: 33049585 DOI: 10.1016/j.coph.2020.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/04/2020] [Indexed: 11/17/2022]
Abstract
Antiretroviral therapy in people living with HIV can achieve potent, long-term suppression of HIV plasma viremia and has increased life expectancy. The central nervous system is infected early after virus acquisition and remains a reservoir for HIV. HIV-associated neurocognitive disorders (HAND) are an end-organ manifestation of HIV infection. The need to address neurological complications caused by HAND is significant as approximately 50% of people living with HIV on suppressive antiretroviral therapy are estimated to have some form of HAND. This review discusses the pathophysiology of HAND, CSF/CNS penetration and clinical pharmacology of antiretrovirals including pharmacokinetic/pharmacodynamic relationships, the persistence of HIV in the brain, and future therapeutic approaches to preserve and improve sustained viral suppression in the brain.
Collapse
Affiliation(s)
- Sean N Avedissian
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, Omaha, NE, United States
| | - Shetty Ravi Dyavar
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, Omaha, NE, United States
| | - Howard S Fox
- Center for Integrative and Translational Neuroscience, Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, Omaha, NE, United States.
| |
Collapse
|
10
|
Otaru S, Niemikoski H, Sarparanta M, Airaksinen AJ. Metabolism of a Bioorthogonal PET Tracer Candidate [ 19F/ 18F]SiFA-Tetrazine in Mouse Liver Microsomes: Biotransformation Pathways and Defluorination Investigated by UHPLC-HRMS. Mol Pharm 2020; 17:3106-3115. [PMID: 32539414 PMCID: PMC7497667 DOI: 10.1021/acs.molpharmaceut.0c00523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Organofluorosilicon based 18F-radiolabeling is an efficient
method for incorporating fluorine-18 into 18F-radiopharmaceuticals
for positron emission tomography (PET) by 19F/18F isotopic exchange (IE). The first PET radiopharmaceutical, 18F-SiFAlin-TATE, radiolabeled with a silicon-based
[18F]fluoride acceptor (SiFA), namely, a para-substituted
di-tert-butyl[18F]fluorosilylbenzene,
has entered clinical trials, and is paving the way for other potential
[18F]SiFA-labeled radiopharmaceuticals for diagnostic use.
In this study, we report the in vitro metabolism
of an oxime-linked SiFA tetrazine (SiFA–Tz), a new PET-radiotracer
candidate, recently evaluated for pretargeted PET imaging and macromolecule
labeling. Metabolism of SiFA–Tz was studied in mouse liver
microsomes (MLM) for elucidating its major biotransformation pathways.
Nontargeted screening by ultrahigh performance liquid chromatography
high-resolution mass spectrometry (UHPLC-HRMS) was utilized for detection
of unknown metabolites. The oxime bond between the SiFA and Tz groups
forms two geometric (E/Z) isomers,
which underwent the same biotransformations, but unexpectedly with
different kinetics. In total, nine proposed metabolites of SiFA–Tz
from phase I and II reactions were detected, five of which were defluorinated
in MLMs, elucidating the metabolic pathway leading to previously reported
defluorination of [18F]SiFA–Tz in vivo. Based on the HRMS studies a biotransformation pathway is proposed:
hydroxylation (+O) to tert-butyl group adjacent to
the silicon, followed by oxidative defluorination (+OH/-F) cleaving
the fluorine off the silicon. Interestingly, eight proposed metabolites
of a reduced dihydrotetrazine analogue, SiFA–H2Tz,
from phase I and II reactions were additionally detected. To the best
of our knowledge, this is the first reported comprehensive investigation
of enzyme mediated metabolic pathway of tetrazines and para-substituted
di-tert-butylfluorosilylbenzene fluoride acceptors,
providing novel structural information on the biotransformation and
fragmentation patterns of radiotracers bearing these structural motifs.
By investigating the metabolism preceding defluorination, structurally
optimized new SiFA compounds can be designed for expanding the portfolio
of efficient 19F/18F isotopic exchange labeling
probes for PET imaging.
Collapse
Affiliation(s)
- Sofia Otaru
- Radiochemistry, Department of Chemistry, University of Helsinki, 00014 Helsinki, Finland
| | - Hanna Niemikoski
- Finnish Institute for Verification of the Chemical Weapons Convention (VERIFIN), Department of Chemistry, University of Helsinki, 00014 Helsinki, Finland
| | - Mirkka Sarparanta
- Radiochemistry, Department of Chemistry, University of Helsinki, 00014 Helsinki, Finland
| | - Anu J Airaksinen
- Radiochemistry, Department of Chemistry, University of Helsinki, 00014 Helsinki, Finland.,Turku PET Centre, Department of Chemistry, University of Turku, 20500 Turku, Finland
| |
Collapse
|
11
|
Kecel-Gunduz S, Budama-Kilinc Y, Cakir-Koc R, Zorlu T, Bicak B, Kokcu Y, E Ozel A, Akyuz S. In Silico design of AVP (4-5) peptide and synthesis, characterization and in vitro activity of chitosan nanoparticles. ACTA ACUST UNITED AC 2020; 28:139-157. [PMID: 31942695 DOI: 10.1007/s40199-019-00325-9] [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: 01/16/2019] [Accepted: 12/23/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Arginine-vasopressin (AVP) is a neuropeptide and provides learning and memory modulation. The AVP (4-5) dipeptide corresponds to the N-terminal fragment of the major vasopressin metabolite AVP (4-9), has a neuroprotective effect and used in the treatment of Alzheimer's and Parkinson's disease. METHODS The main objective of the present study is to evaluate the molecular mechanism of AVP (4-5) dipeptide and to develop and synthesize chitosan nanoparticle formulation using modified version of ionic gelation method, to increase drug effectiveness. For peptide loaded chitosan nanoparticles, the synthesized experiment medium was simulated for the first time by molecular dynamics method and used to determine the stability of the peptide, and the binding mechanism to protein (HSP70) was also investigated by molecular docking calculations. A potential pharmacologically features of the peptide was also characterized by ADME (Absorption, Distribution, Metabolism and Excretion) analysis. The characterization, in vitro release study, encapsulation efficiency and loading capacity of the peptide loaded chitosan nanoparticles (CS NPs) were performed by Dynamic Light Scattering (DLS), UV-vis absorption (UV), Scanning Electron Microscopy (SEM), Fourier transform infrared (FT-IR) spectroscopy techniques. Additionally, in vitro cytotoxicity of the peptide on human neuroblastoma cells (SH-SY5Y) was examined with XTT assay and the statistical analysis was evaluated. RESULTS The results showed that; hydrodynamic size, zeta potential and polydispersity index (PdI) of the peptide-loaded CS NPs were 167.6 nm, +13.2 mV, and 0.211, respectively. In vitro release study of the peptide-loaded CS NPs showed that 17.23% of the AVP (4-5)-NH2 peptide was released in the first day, while 61.13% of AVP (4-5)-NH2 peptide was released in the end of the 10th day. The encapsulation efficiency and loading capacity were 99% and 10%, respectively. According to the obtained results from XTT assay, toxicity on SHSY-5Y cells in the concentration from 0.01 μg/μL to 30 μg/μL were evaluated and no toxicity was observed. Also, neuroprotective effect was showed against H2O2 treatment. CONCLUSION The experimental medium of peptide-loaded chitosan nanoparticles was created for the first time with in silico system and the stability of the peptide in this medium was carried out by molecular dynamics studies. The binding sites of the peptide with the HSP70 protein were determined by molecular docking analysis. The size and morphology of the prepared NPs capable of crossing the blood-brain barrier (BBB) were monitored using DLS and SEM analyses, and the encapsulation efficiency and loading capacity were successfully performed with UV Analysis. In vitro release studies and in vitro cytotoxicity analysis on SHSY-5Y cell lines of the peptide were conducted for the first time. Grapical abstract.
Collapse
Affiliation(s)
- Serda Kecel-Gunduz
- Physics Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey.
| | - Yasemin Budama-Kilinc
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, 34220, Istanbul, Turkey
| | - Rabia Cakir-Koc
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, 34220, Istanbul, Turkey
| | - Tolga Zorlu
- Graduate School of Natural and Applied Science, Yildiz Technical University, 34220, Istanbul, Turkey.,Department of Physical Chemistry and EMaS, Universitat Rovira i Virgili, 43007, Tarragona, Spain
| | - Bilge Bicak
- Physics Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey.,Institute of Graduate Studies in Sciences, Istanbul University, 34452, Istanbul, Turkey
| | - Yagmur Kokcu
- Institute of Graduate Studies in Sciences, Istanbul University, 34452, Istanbul, Turkey
| | - Aysen E Ozel
- Physics Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey
| | - Sevim Akyuz
- Physics Department, Science and Letters Faculty, Istanbul Kultur University, Atakoy Campus, Bakirkoy, 34156, Istanbul, Turkey
| |
Collapse
|
12
|
Liu F, Dai S, Feng D, Peng X, Qin Z, Kearns AC, Huang W, Chen Y, Ergün S, Wang H, Rappaport J, Bryda EC, Chandrasekhar A, Aktas B, Hu H, Chang SL, Gao B, Qin X. Versatile cell ablation tools and their applications to study loss of cell functions. Cell Mol Life Sci 2019; 76:4725-4743. [PMID: 31359086 PMCID: PMC6858955 DOI: 10.1007/s00018-019-03243-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/22/2022]
Abstract
Targeted cell ablation is a powerful approach for studying the role of specific cell populations in a variety of organotypic functions, including cell differentiation, and organ generation and regeneration. Emerging tools for permanently or conditionally ablating targeted cell populations and transiently inhibiting neuronal activities exhibit a diversity of application and utility. Each tool has distinct features, and none can be universally applied to study different cell types in various tissue compartments. Although these tools have been developed for over 30 years, they require additional improvement. Currently, there is no consensus on how to select the tools to answer the specific scientific questions of interest. Selecting the appropriate cell ablation technique to study the function of a targeted cell population is less straightforward than selecting the method to study a gene's functions. In this review, we discuss the features of the various tools for targeted cell ablation and provide recommendations for optimal application of specific approaches.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shen Dai
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiao Peng
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Zhongnan Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Alison C Kearns
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Yong Chen
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, 261053, Weifang, People's Republic of China
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximillan University, 97070, Wurzburg, Germany
| | - Hong Wang
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jay Rappaport
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Elizabeth C Bryda
- Rat Resource and Research Center, University of Missouri, 4011 Discovery Drive, Columbia, MO, 65201, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences, 340D Life Sciences Center, University of Missouri, 1201 Rollins St, Columbia, MO, USA
| | - Bertal Aktas
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
13
|
Colom M, Vidal B, Zimmer L. Is There a Role for GPCR Agonist Radiotracers in PET Neuroimaging? Front Mol Neurosci 2019; 12:255. [PMID: 31680859 PMCID: PMC6813225 DOI: 10.3389/fnmol.2019.00255] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/02/2019] [Indexed: 12/30/2022] Open
Abstract
Positron emission tomography (PET) is a molecular imaging modality that enables in vivo exploration of metabolic processes and especially the pharmacology of neuroreceptors. G protein-coupled receptors (GPCRs) play an important role in numerous pathophysiologic disorders of the central nervous system. Thus, they are targets of choice in PET imaging to bring proof concept of change in density in pathological conditions or in pharmacological challenge. At present, most radiotracers are antagonist ligands. In vitro data suggest that properties differ between GPCR agonists and antagonists: antagonists bind to receptors with a single affinity, whereas agonists are characterized by two different affinities: high affinity for receptors that undergo functional coupling to G-proteins, and low affinity for those that are not coupled. In this context, agonist radiotracers may be useful tools to give functional images of GPCRs in the brain, with high sensitivity to neurotransmitter release. Here, we review all existing PET radiotracers used from animals to humans and their role for understanding the ligand-receptor paradigm of GPCR in comparison with corresponding antagonist radiotracers.
Collapse
Affiliation(s)
- Matthieu Colom
- Lyon Neuroscience Research Center, INSERM, CNRS, Université de Lyon, Lyon, France.,CERMEP, Hospices Civils de Lyon, Bron, France
| | - Benjamin Vidal
- Lyon Neuroscience Research Center, INSERM, CNRS, Université de Lyon, Lyon, France
| | - Luc Zimmer
- Lyon Neuroscience Research Center, INSERM, CNRS, Université de Lyon, Lyon, France.,CERMEP, Hospices Civils de Lyon, Bron, France.,Institut National des Sciences et Techniques Nucléaires, CEA Saclay, Gif-sur-Yvette, France
| |
Collapse
|
14
|
Human biodistribution and radiation dosimetry of the 5-HT 2A receptor agonist Cimbi-36 labeled with carbon-11 in two positions. EJNMMI Res 2019; 9:71. [PMID: 31367837 PMCID: PMC6669221 DOI: 10.1186/s13550-019-0527-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/20/2019] [Indexed: 12/17/2022] Open
Abstract
Background Cimbi-36 can be 11C-labeled to form an agonist radioligand used for positron emission tomography (PET) imaging of the 5-HT2A receptor in the brain. In its non-labeled form (25B-NBOMe), it is used as a recreational drug that can lead to severe adverse effects, in some cases, with fatal outcome. We investigated human biodistribution and radiation dosimetry of the radioligand with two different radiolabeling positions. Seven healthy volunteers underwent dynamic 120-min whole-body PET scans (injection of 581 ± 16 MBq, n = 5 for 11C-Cimbi-36; 593 ± 14 MBq, n = 2 for 11C-Cimbi-36_5). Time-integrated activity coefficients (TIACs) from time-activity curves (TACs) of selected organs were used as input into the OLINDA/EXM software to obtain dosimetry information for both 11C-labeling positions of Cimbi-36. Results The effective dose was only slightly higher for 11C-Cimbi-36 (5.5 μSv/MBq) than for 11C-Cimbi-36_5 (5.3 μSv/MBq). Standard uptake value (SUV) curves showed higher uptake of 11C-Cimbi-36 in the pancreas, small intestines, liver, kidney, gallbladder, and urinary bladder compared with 11C-Cimbi-36_5, reflecting differences in radiometabolism for the two radioligands. Variability in uptake in excretory organs for 11C-Cimbi-36 points to inter-individual differences with regard to metabolic rate and route. Surprisingly, moderate uptake was found in brown adipose tissue (BAT) in four subjects, possibly representing specific 5-HT2A/2C receptor binding. Conclusion The low effective dose of 5.5 μSv/MBq allows for the injection of up to 1.8 GBq for healthy volunteers per study (equivalent to 3 scans if injecting 600 MBq) and still stay below the international guidelines of 10 mSv, making 11C-Cimbi-36 eligible for studies involving a series of PET scans in a single subject. The biodistribution of Cimbi-36 (and its metabolites) may also help to shed light on the toxic effects of 25B-NBOMe when used in pharmacological doses in recreational settings. Electronic supplementary material The online version of this article (10.1186/s13550-019-0527-4) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Canal CE. Serotonergic Psychedelics: Experimental Approaches for Assessing Mechanisms of Action. Handb Exp Pharmacol 2019; 252:227-260. [PMID: 29532180 PMCID: PMC6136989 DOI: 10.1007/164_2018_107] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent, well-controlled - albeit small-scale - clinical trials show that serotonergic psychedelics, including psilocybin and lysergic acid diethylamide, possess great promise for treating psychiatric disorders, including treatment-resistant depression. Additionally, fresh results from a deluge of clinical neuroimaging studies are unveiling the dynamic effects of serotonergic psychedelics on functional activity within, and connectivity across, discrete neural systems. These observations have led to testable hypotheses regarding neural processing mechanisms that contribute to psychedelic effects and therapeutic benefits. Despite these advances and a plethora of preclinical and clinical observations supporting a central role for brain serotonin 5-HT2A receptors in producing serotonergic psychedelic effects, lingering and new questions about mechanisms abound. These chiefly pertain to molecular neuropharmacology. This chapter is devoted to illuminating and discussing such questions in the context of preclinical experimental approaches for studying mechanisms of action of serotonergic psychedelics, classic and new.
Collapse
Affiliation(s)
- Clinton E Canal
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA.
| |
Collapse
|
16
|
Psychedelic effects of psilocybin correlate with serotonin 2A receptor occupancy and plasma psilocin levels. Neuropsychopharmacology 2019; 44:1328-1334. [PMID: 30685771 PMCID: PMC6785028 DOI: 10.1038/s41386-019-0324-9] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/17/2018] [Accepted: 12/26/2018] [Indexed: 11/08/2022]
Abstract
The main psychedelic component of magic mushrooms is psilocybin, which shows promise as a treatment for depression and other mental disorders. Psychedelic effects are believed to emerge through stimulation of serotonin 2A receptors (5-HT2ARs) by psilocybin's active metabolite, psilocin. We here report for the first time the relationship between intensity of psychedelic effects, cerebral 5-HT2AR occupancy and plasma levels of psilocin in humans. Eight healthy volunteers underwent positron emission tomography (PET) scans with the 5-HT2AR agonist radioligand [11C]Cimbi-36: one at baseline and one or two additional scans on the same day after a single oral intake of psilocybin (3-30 mg). 5-HT2AR occupancy was calculated as the percent change in cerebral 5-HT2AR binding relative to baseline. Subjective psychedelic intensity and plasma psilocin levels were measured during the scans. Relations between subjective intensity, 5-HT2AR occupancy, and plasma psilocin levels were modeled using non-linear regression. Psilocybin intake resulted in dose-related 5-HT2AR occupancies up to 72%; plasma psilocin levels and 5-HT2AR occupancy conformed to a single-site binding model. Subjective intensity was correlated with both 5-HT2AR occupancy and psilocin levels as well as questionnaire scores. We report for the first time that intake of psilocybin leads to significant 5-HT2AR occupancy in the human brain, and that both psilocin plasma levels and 5-HT2AR occupancy are closely associated with subjective intensity ratings, strongly supporting that stimulation of 5-HT2AR is a key determinant for the psychedelic experience. Important for clinical studies, psilocin time-concentration curves varied but psilocin levels were closely associated with psychedelic experience.
Collapse
|
17
|
Shalgunov V, van Waarde A, Booij J, Michel MC, Dierckx RAJO, Elsinga PH. Hunting for the high-affinity state of G-protein-coupled receptors with agonist tracers: Theoretical and practical considerations for positron emission tomography imaging. Med Res Rev 2018; 39:1014-1052. [PMID: 30450619 PMCID: PMC6587759 DOI: 10.1002/med.21552] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/02/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
Abstract
The concept of the high‐affinity state postulates that a certain subset of G‐protein‐coupled receptors is primarily responsible for receptor signaling in the living brain. Assessing the abundance of this subset is thus potentially highly relevant for studies concerning the responses of neurotransmission to pharmacological or physiological stimuli and the dysregulation of neurotransmission in neurological or psychiatric disorders. The high‐affinity state is preferentially recognized by agonists in vitro. For this reason, agonist tracers have been developed as tools for the noninvasive imaging of the high‐affinity state with positron emission tomography (PET). This review provides an overview of agonist tracers that have been developed for PET imaging of the brain, and the experimental paradigms that have been developed for the estimation of the relative abundance of receptors configured in the high‐affinity state. Agonist tracers appear to be more sensitive to endogenous neurotransmitter challenge than antagonists, as was originally expected. However, other expectations regarding agonist tracers have not been fulfilled. Potential reasons for difficulties in detecting the high‐affinity state in vivo are discussed.
Collapse
Affiliation(s)
- Vladimir Shalgunov
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine, Ghent University, University Hospital, Ghent, Belgium
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|