1
|
Borrega-Roman L, Hoare BL, Kosar M, Sarott RC, Patej KJ, Bouma J, Scott-Dennis M, Koers EJ, Gazzi T, Mach L, Barrondo S, Sallés J, Guba W, Kusznir E, Nazaré M, Rufer AC, Grether U, Heitman LH, Carreira EM, Sykes DA, Veprintsev DB. A universal cannabinoid CB1 and CB2 receptor TR-FRET kinetic ligand-binding assay. Front Pharmacol 2025; 16:1469986. [PMID: 40271066 PMCID: PMC12015242 DOI: 10.3389/fphar.2025.1469986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/11/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction The kinetics of ligand binding to G protein-coupled receptors (GPCRs) is an important optimization parameter in drug discovery. Traditional radioligand assays are labor-intensive, preventing their application at the early stages of drug discovery. Fluorescence-based assays offer several advantages, including a possibility to develop a homogeneous format, continuous data collection, and higher throughput. This study sought to develop a fluorescence-based binding assay to investigate ligand-binding kinetics at human cannabinoid type 1 and 2 receptors (CB1R and CB2R). Methods We synthesized D77, a novel tracer derived from the non-selective cannabinoid Δ8-THC. Using time-resolved Förster resonance energy transfer (TR-FRET), we developed an assay to study ligand-binding kinetics at physiological temperatures. For CB1R, we truncated the first 90 amino acids of its flexible N-terminal domain to reduce the FRET distance between the terbium cryptate (donor) and the fluorescent ligand (acceptor). The full-length CB2R construct was functional without modification due to its shorter N-terminus. The Motulsky-Mahan competition binding model was used to analyze the binding kinetics of the endocannabinoids and several other non-fluorescent ligands. Results The D77 tracer showed nanomolar-range affinity for truncated CB1R (CB1R91-472) and full-length CB2R (CB2R1-360), displaying competitive binding with orthosteric ligands. D77 exhibited rapid dissociation kinetics from both CB1R and CB2R, which were similar to the fastest dissociating reference compounds. This was critical for accurately determining the on- and off-rates of the fastest dissociating compounds. Using D77, we measured the kinetic binding properties of various CB1R and CB2R agonists and antagonists at physiological temperature and sodium ion concentration. Discussion The k on values for molecules binding to CB1R varied by three orders of magnitude, from the slowest (HU308) to the fastest (rimonabant). A strong correlation between k on and affinity was observed for compounds binding to CB1R, indicating that the association rate primarily determines their affinity for CB1R. Unlike CB1R, a stronger correlation was found between the dissociation rate constant k off and the affinity for CB2R, suggesting that both k on and k off dictate the overall affinity for CB2R. Exploring the kinetic parameters of cannabinoid drug candidates could help drug development programs targeting these receptors.
Collapse
Affiliation(s)
- Leire Borrega-Roman
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
| | - Bradley L. Hoare
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Miroslav Kosar
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Roman C. Sarott
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Kacper J. Patej
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Jara Bouma
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University and Oncode Institute, Leiden, Netherlands
| | - Morgan Scott-Dennis
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Eline J. Koers
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Thais Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Campus BerlinBuch, Berlin, Germany
| | - Leonard Mach
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Campus BerlinBuch, Berlin, Germany
| | - Sergio Barrondo
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Joan Sallés
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Wolfgang Guba
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Eric Kusznir
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Campus BerlinBuch, Berlin, Germany
| | - Arne C. Rufer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Uwe Grether
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Laura H. Heitman
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden University and Oncode Institute, Leiden, Netherlands
| | - Erick M. Carreira
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - David A. Sykes
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| | - Dmitry B. Veprintsev
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom
| |
Collapse
|
2
|
van Wamelen DJ, Martin NH, Makos O, Badenoch J, Valera-Bermejo JM, Hartmann M, Cristales AR, Wood TC, Veronese M, Moretto M, Zelaya F, dell'Acqua F, O'Daly O, Lythgoe DJ, Ginestet C, Turkheimer F, Palasits N, Mrzljak L, Warner JH, Rabiner EA, Gunn R, Tabrizi SJ, Sampaio C, Wood A, Williams SC. Study protocol for the iMarkHD study in individuals with Huntington's disease. J Huntingtons Dis 2024; 13:479-489. [PMID: 39973385 DOI: 10.1177/18796397241288165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background: Huntington's disease (HD) is still often defined by the onset of motor symptoms, inversely associated with the size of the CAG repeat expansion in the huntingtin gene. Although the cause of HD is known, much remains unknown about mechanisms underlying clinical symptom development, disease progression, and specific clinical subtypes/endophenotypes. Objective: In the iMarkHD study, we aim to investigate four discrete molecular positron emission tomography (PET) tracers and magnetic resonance imaging (MRI) markers as biomarkers for disease and symptom progression. Methods: Following MRI optimization in five healthy volunteers (cohort 1), we aim to recruit 108 participants of whom 72 are people with HD (PwHD) and 36 healthy volunteers (cohort 2). Pending interim analysis, these numbers could increase to 96 PwHD and 48 healthy controls. Participants will complete a total of 10 study visits, consisting of a screening visit followed by a clinical and MRI visit and PET visits at baseline, year 1, and year 2. PET targets include the cannabinoid 1, histamine 3, and serotonin 2A receptors, and phosphodiesterase 10A, whereas MRI will be multimodal, including, but not limited to, the assessment of cerebral blood flow, functional connectivity, and brain iron. Results: Recruitment is currently active and started in September 2022. Conclusions: By combining PET and multi-modal MRI assessments we expect to provide a comprehensive examination of the molecular, functional, and structural framework of HD progression. As such, the iMarkHD study will provide a solid base for the identification of treatment targets and novel outcome measures for future clinical trials.
Collapse
Affiliation(s)
- Daniel J van Wamelen
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Centre of Expertise for Parkinson & Movement Disorders, Nijmegen, the Netherlands
| | - Naomi H Martin
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Orsolya Makos
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - James Badenoch
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Jose Manuel Valera-Bermejo
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Monika Hartmann
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Alay Rangel Cristales
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Tobias C Wood
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Mattia Veronese
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Manuela Moretto
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Fernando Zelaya
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Flavio dell'Acqua
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Owen O'Daly
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - David J Lythgoe
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Cedric Ginestet
- Institute of Psychiatry, Psychology & Neuroscience, Department of Biostatistics & Health Informatics, King's College London, London, UK
| | - Federico Turkheimer
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| | - Nikki Palasits
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | - Ladislav Mrzljak
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
- Takeda Pharmaceuticals, Cambridge, MA, USA
| | - John H Warner
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | | | - Roger Gunn
- Invicro, A Konica Minolta Company, London, UK
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, UK Dementia Research Institute, Department of Neurodegenerative Diseases, University College London, London, UK
| | - Cristina Sampaio
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | - Andrew Wood
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Princeton, NJ, USA
| | - Steven Cr Williams
- Institute of Psychiatry, Psychology & Neuroscience, Department of Neuroimaging, King's College London, London, UK
| |
Collapse
|
3
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Sexton M, Garcia JM, Jatoi A, Clark CS, Wallace MS. The Management of Cancer Symptoms and Treatment-Induced Side Effects With Cannabis or Cannabinoids. J Natl Cancer Inst Monogr 2021; 2021:86-98. [PMID: 34850897 DOI: 10.1093/jncimonographs/lgab011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 01/06/2023] Open
Abstract
Cannabis and cannabinoids are increasingly being accessed and used by patients with advanced cancer for various symptoms and general quality of life. Specific symptoms of pain, nausea and vomiting, loss of appetite and cachexia, anxiety, sleep disturbance, and medical trauma are among those that have prompted patients with cancer to use cannabis. This conference report from the National Cancer Institute's "Cannabis, Cannabinoid and Cancer Research Symposium" on the topic of "Cancer Symptom/Treatment Side Effect Management" is an expert perspective of cannabis intervention for cancer and cancer treatment-related symptoms. The purpose of the symposium was to identify research gaps, describe the need for high-quality randomized prospective studies of medical cannabis for palliative care in patients with cancer, and evaluate the impact of medical cannabis on cancer survivors' quality of life. Further, education of clinicians and affiliated health-care providers in guiding cancer patients in using cannabis for cancer care would benefit patients. Together, these steps will further aid in refining the use of cannabis and cannabinoids for symptom palliation and improve safety and efficacy for patients.
Collapse
Affiliation(s)
- Michelle Sexton
- Department of Anesthesiology, Division of Pain Management, University of California, San Diego, CA, USA
| | - Jose M Garcia
- Department of Medicine, Division of Geriatrics, University of Washington and Geriatric Research Education and Clinical Center, Puget Sound Veterans Administration Health Care System, Seattle, WA, USA
| | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Carey S Clark
- Department of Nursing, Pacific College of Health and Science, San Diego, CA, USA
| | - Mark S Wallace
- Department of Anesthesiology, Division of Pain Management, University of California, San Diego, CA, USA
| |
Collapse
|
5
|
Spindle TR, Kuwabara H, Eversole A, Nandi A, Vandrey R, Antoine DG, Umbricht A, Guarda AS, Wong DF, Weerts EM. Brain imaging of cannabinoid type I (CB 1 ) receptors in women with cannabis use disorder and male and female healthy controls. Addict Biol 2021; 26:e13061. [PMID: 34028926 PMCID: PMC8516687 DOI: 10.1111/adb.13061] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/19/2022]
Abstract
Cannabis effects are predominantly mediated by pharmacological actions on cannabinoid type 1 (CB1 ) receptors. Prior positron emission tomography (PET) studies in individuals who use cannabis included almost exclusively males. PET studies in females are needed because there are sex differences in cannabis effects, progression to cannabis use disorder (CUD), and withdrawal symptom severity. Females with CUD (N = 10) completed two double-blind cannabis smoking sessions (Session 1: placebo; Session 2: active), and acute cannabis effects were assessed. After Session 2, participants underwent 3 days of monitored cannabis abstinence; mood, craving, and withdrawal symptoms were assessed and a PET scan (radiotracer: [11 C]OMAR) followed. [11 C]OMAR Distribution volume (VT ) from these participants was compared with VT of age/BMI-similar female non-users of cannabis ("healthy controls"; N = 10). VT was also compared between female and male healthy controls (N = 7). Females with CUD displayed significantly lower VT than female healthy controls in specific brain regions (hippocampus, amygdala, cingulate, and insula). Amygdala VT was negatively correlated with mood changes (anger/hostility) during abstinence, but VT was not correlated with other withdrawal symptoms or cannabis effects. Among healthy controls, females had significantly higher VT than males in all brain regions examined. Chronic cannabis use appears to foster downregulation of CB1 receptors in women, as observed previously in men, and there are inherent sex differences in CB1 availability. Future studies should elucidate the time course of CB1 downregulation among females who use cannabis and examine the relation between CB1 availability and cannabis effects among other populations (e.g., infrequent users; medicinal users).
Collapse
Affiliation(s)
- Tory R. Spindle
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Hiroto Kuwabara
- Division of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline Street, Baltimore, MD, 21287, USA
| | - Alisha Eversole
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Ayon Nandi
- Division of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline Street, Baltimore, MD, 21287, USA
| | - Ryan Vandrey
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Denis G. Antoine
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Annie Umbricht
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Angela S. Guarda
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Dean F. Wong
- Division of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline Street, Baltimore, MD, 21287, USA
| | - Elise M. Weerts
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| |
Collapse
|
6
|
Ma L, Wu S, Zhang K, Tian M, Zhang H. Progress on the application of positron emission tomography imaging of cannabinoid type 1 receptor in neuropsychiatric diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:666-673. [PMID: 34986538 PMCID: PMC8732249 DOI: 10.3724/zdxbyxb-2021-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cannabinoid type 1 receptor (CB1R), as the major member of the endocannabinoid system, is among the most abundant receptors expressed in the central nervous system. CB1R is mainly located on the axon terminals of presynaptic neurons and participate in the modulation of neuronal excitability and synaptic plasticity, playing an important role in the pathogenesis of various neuropsychiatric diseases. In recent years, the consistent development of CB1R radioligands and the maturity of molecular imaging techniques, particularly positron emission tomography (PET) may help to visualize the expression and distribution of CB1R in central nervous system . At present, CB1R PET imaging can effectively evaluate the changes of CB1R levels in neuropsychiatric diseases such as Huntington's disease and schizophrenia, and its correlation with the disease severity, therefore providing new insights for the diagnosis and treatment of neuropsychiatric diseases. This article reviews the application of CB1R PET imaging in Alzheimer's disease, Parkinson's disease, Huntington's disease, schizophrenia, post-traumatic stress disorder, cannabis use disorder and depression.
Collapse
Affiliation(s)
- Lijuan Ma
- 4. College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Shuang Wu
- 4. College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Kai Zhang
- 4. College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Mei Tian
- 4. College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Hong Zhang
- 4. College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
7
|
Takamura Y, Kakuta H. In Vivo Receptor Visualization and Evaluation of Receptor Occupancy with Positron Emission Tomography. J Med Chem 2021; 64:5226-5251. [PMID: 33905258 DOI: 10.1021/acs.jmedchem.0c01714] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Positron emission tomography (PET) is useful for noninvasive in vivo visualization of disease-related receptors, for evaluation of receptor occupancy to determine an appropriate drug dosage, and for proof-of-concept of drug candidates in translational research. For these purposes, the specificity of the PET tracer for the target receptor is critical. Here, we review work in this area, focusing on the chemical structures of reported PET tracers, their Ki/Kd values, and the physical properties relevant to target receptor selectivity. Among these physical properties, such as cLogP, cLogD, molecular weight, topological polar surface area, number of hydrogen bond donors, and pKa, we focus especially on LogD and LogP as important physical properties that can be easily compared across a range of studies. We discuss the success of PET tracers in evaluating receptor occupancy and consider likely future developments in the field.
Collapse
Affiliation(s)
- Yuta Takamura
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
8
|
Hou L, Rong J, Haider A, Ogasawara D, Varlow C, Schafroth MA, Mu L, Gan J, Xu H, Fowler CJ, Zhang MR, Vasdev N, Ametamey S, Cravatt BF, Wang L, Liang SH. Positron Emission Tomography Imaging of the Endocannabinoid System: Opportunities and Challenges in Radiotracer Development. J Med Chem 2021; 64:123-149. [PMID: 33379862 PMCID: PMC7877880 DOI: 10.1021/acs.jmedchem.0c01459] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The endocannabinoid system (ECS) is involved in a wide range of biological functions and comprises cannabinoid receptors and enzymes responsible for endocannabinoid synthesis and degradation. Over the past 2 decades, significant advances toward developing drugs and positron emission tomography (PET) tracers targeting different components of the ECS have been made. Herein, we summarized the recent development of PET tracers for imaging cannabinoid receptors 1 (CB1R) and 2 (CB2R) as well as the key enzymes monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), particularly focusing on PET neuroimaging applications. State-of-the-art PET tracers for the ECS will be reviewed including their chemical design, pharmacological properties, radiolabeling, as well as preclinical and human PET imaging. In addition, this review addresses the current challenges for ECS PET biomarker development and highlights the important role of PET ligands to study disease pathophysiology as well as to facilitate drug discovery.
Collapse
Affiliation(s)
- Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Daisuke Ogasawara
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health & Department of Psychiatry/Institute of Medical Science, University of Toronto, 250 College St., Toronto, M5T 1R8, ON., Canada
| | - Michael A. Schafroth
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Linjing Mu
- Center for Radiopharmaceutical Sciences ETH, PSI and USZ, Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Jiefeng Gan
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Christopher J. Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health & Department of Psychiatry/Institute of Medical Science, University of Toronto, 250 College St., Toronto, M5T 1R8, ON., Canada
| | - Simon Ametamey
- Center for Radiopharmaceutical Sciences ETH, PSI and USZ, Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Benjamin F. Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
9
|
Terry GE, Raymont V, Horti AG. PET Imaging of the Endocannabinoid System. PET AND SPECT OF NEUROBIOLOGICAL SYSTEMS 2021:319-426. [DOI: 10.1007/978-3-030-53176-8_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
D'Elia A, Schiavi S, Soluri A, Massari R, Soluri A, Trezza V. Role of Nuclear Imaging to Understand the Neural Substrates of Brain Disorders in Laboratory Animals: Current Status and Future Prospects. Front Behav Neurosci 2020; 14:596509. [PMID: 33362486 PMCID: PMC7759612 DOI: 10.3389/fnbeh.2020.596509] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Molecular imaging, which allows the real-time visualization, characterization and measurement of biological processes, is becoming increasingly used in neuroscience research. Scintigraphy techniques such as single photon emission computed tomography (SPECT) and positron emission tomography (PET) provide qualitative and quantitative measurement of brain activity in both physiological and pathological states. Laboratory animals, and rodents in particular, are essential in neuroscience research, providing plenty of models of brain disorders. The development of innovative high-resolution small animal imaging systems together with their radiotracers pave the way to the study of brain functioning and neurotransmitter release during behavioral tasks in rodents. The assessment of local changes in the release of neurotransmitters associated with the performance of a given behavioral task is a turning point for the development of new potential drugs for psychiatric and neurological disorders. This review addresses the role of SPECT and PET small animal imaging systems for a better understanding of brain functioning in health and disease states. Brain imaging in rodent models faces a series of challenges since it acts within the boundaries of current imaging in terms of sensitivity and spatial resolution. Several topics are discussed, including technical considerations regarding the strengths and weaknesses of both technologies. Moreover, the application of some of the radioligands developed for small animal nuclear imaging studies is discussed. Then, we examine the changes in metabolic and neurotransmitter activity in various brain areas during task-induced neural activation with special regard to the imaging of opioid, dopaminergic and cannabinoid receptors. Finally, we discuss the current status providing future perspectives on the most innovative imaging techniques in small laboratory animals. The challenges and solutions discussed here might be useful to better understand brain functioning allowing the translation of preclinical results into clinical applications.
Collapse
Affiliation(s)
- Annunziata D'Elia
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| | - Sara Schiavi
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| | - Andrea Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Roberto Massari
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Alessandro Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Viviana Trezza
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| |
Collapse
|
11
|
Ghosh KK, Padmanabhan P, Yang CT, Mishra S, Halldin C, Gulyás B. Dealing with PET radiometabolites. EJNMMI Res 2020; 10:109. [PMID: 32997213 PMCID: PMC7770856 DOI: 10.1186/s13550-020-00692-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
Abstract Positron emission tomography (PET) offers the study of biochemical,
physiological, and pharmacological functions at a cellular and molecular level.
The performance of a PET study mostly depends on the used radiotracer of
interest. However, the development of a novel PET tracer is very difficult, as
it is required to fulfill a lot of important criteria. PET radiotracers usually
encounter different chemical modifications including redox reaction, hydrolysis,
decarboxylation, and various conjugation processes within living organisms. Due
to this biotransformation, different chemical entities are produced, and the
amount of the parent radiotracer is declined. Consequently, the signal measured
by the PET scanner indicates the entire amount of radioactivity deposited in the
tissue; however, it does not offer any indication about the chemical disposition
of the parent radiotracer itself. From a radiopharmaceutical perspective, it is
necessary to quantify the parent radiotracer’s fraction present in the tissue.
Hence, the identification of radiometabolites of the radiotracers is vital for
PET imaging. There are mainly two reasons for the chemical identification of PET
radiometabolites: firstly, to determine the amount of parent radiotracers in
plasma, and secondly, to rule out (if a radiometabolite enters the brain) or
correct any radiometabolite accumulation in peripheral tissue. Besides,
radiometabolite formations of the tracer might be of concern for the PET study,
as the radiometabolic products may display considerably contrasting distribution
patterns inside the body when compared with the radiotracer itself. Therefore,
necessary information is needed about these biochemical transformations to
understand the distribution of radioactivity throughout the body. Various
published review articles on PET radiometabolites mainly focus on the sample
preparation techniques and recently available technology to improve the
radiometabolite analysis process. This article essentially summarizes the
chemical and structural identity of the radiometabolites of various radiotracers
including [11C]PBB3,
[11C]flumazenil,
[18F]FEPE2I, [11C]PBR28,
[11C]MADAM, and
(+)[18F]flubatine. Besides, the importance of
radiometabolite analysis in PET imaging is also briefly summarized. Moreover,
this review also highlights how a slight chemical modification could reduce the
formation of radiometabolites, which could interfere with the results of PET
imaging. Graphical abstract ![]()
Collapse
Affiliation(s)
- Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore.
| | - Chang-Tong Yang
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore.,Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore.,Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Sachin Mishra
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Christer Halldin
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore.,Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, SE-171 76, Stockholm, Sweden
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore. .,Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
12
|
Liu GL, Cui YF, Lu C, Zhao P. Ketamine a dissociative anesthetic: Neurobiology and biomolecular exploration in depression. Chem Biol Interact 2020; 319:109006. [DOI: 10.1016/j.cbi.2020.109006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 10/25/2022]
|
13
|
Shollenbarger S, Thomas AM, Wade NE, Gruber SA, Tapert SF, Filbey FM, Lisdahl KM. Intrinsic Frontolimbic Connectivity and Mood Symptoms in Young Adult Cannabis Users. Front Public Health 2019; 7:311. [PMID: 31737591 PMCID: PMC6838025 DOI: 10.3389/fpubh.2019.00311] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/14/2019] [Indexed: 11/25/2022] Open
Abstract
Objective: The endocannbinoid system and cannabis exposure has been implicated in emotional processing. The current study examined whether regular cannabis users demonstrated abnormal intrinsic (a.k.a. resting state) frontolimbic connectivity compared to non-users. A secondary aim examined the relationship between cannabis group connectivity differences and self-reported mood and affect symptoms. Method: Participants included 79 cannabis-using and 80 non-using control emerging adults (ages of 18–30), balanced for gender, reading ability, and age. Standard multiple regressions were used to predict if cannabis group status was associated with frontolimbic connectivity after controlling for site, past month alcohol and nicotine use, and days of abstinence from cannabis. Results: After controlling for research site, past month alcohol and nicotine use, and days of abstinence from cannabis, cannabis users demonstrated significantly greater connectivity between left rACC and the following: right rACC (p = 0.001; corrected p = 0.05; f2 = 0.55), left amygdala (p = 0.03; corrected p = 0.47; f2 = 0.17), and left insula (p = 0.03; corrected p = 0.47; f2 = 0.16). Among cannabis users, greater bilateral rACC connectivity was significantly associated with greater subthreshold depressive symptoms (p = 0.02). Conclusions: Cannabis using young adults demonstrated greater connectivity within frontolimbic regions compared to controls. In cannabis users, greater bilateral rACC intrinsic connectivity was associated with greater levels of subthreshold depression symptoms. Current findings suggest that regular cannabis use during adolescence is associated with abnormal frontolimbic connectivity, especially in cognitive control and emotion regulation regions.
Collapse
Affiliation(s)
- Skyler Shollenbarger
- Psychology Department, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Alicia M Thomas
- Psychology Department, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Natasha E Wade
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
| | - Staci A Gruber
- Imaging Center, McLean Hospital, Belmont, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Susan F Tapert
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
| | - Francesca M Filbey
- Bert Moore Chair in BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, United States
| | - Krista M Lisdahl
- Psychology Department, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| |
Collapse
|
14
|
Taddei C, Pike VW. [ 11C]Carbon monoxide: advances in production and application to PET radiotracer development over the past 15 years. EJNMMI Radiopharm Chem 2019; 4:25. [PMID: 31659516 PMCID: PMC6751244 DOI: 10.1186/s41181-019-0073-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/22/2019] [Indexed: 12/18/2022] Open
Abstract
[11C]Carbon monoxide is an appealing synthon for introducing carbon-11 at a carbonyl position (C=O) in a wide variety of chemotypes (e.g., amides, ketones, acids, esters, and ureas). The prevalence of the carbonyl group in drug molecules and the present-day broad versatility of carbonylation reactions have led to an upsurge in the production of this synthon and in its application to PET radiotracer development. This review focuses on the major advances of the past 15 years.
Collapse
Affiliation(s)
- Carlotta Taddei
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Rm B3C342, Bethesda, MD, 20892-1003, USA.
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Rm B3C342, Bethesda, MD, 20892-1003, USA
| |
Collapse
|
15
|
Valenta I, Pacher P, Dilsizian V, Schindler TH. Novel Myocardial PET/CT Receptor Imaging and Potential Therapeutic Targets. Curr Cardiol Rep 2019; 21:55. [PMID: 31104205 DOI: 10.1007/s11886-019-1148-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF THE REVIEW Activation of myocardial cannabinoid type 1 receptors (CB1-R) and/or angiotensin II type 1 receptors (AT1-R) likely plays an important mechanistic role in determining the left-ventricular remodeling process in systolic heart failure. We provide an overview on novel radiotracer probes and positron emission tomography (PET)/computed tomography (CT) imaging to noninvasively probe the expression of myocardial CB1-R and/or AT1-R. RECENT FINDINGS Recent translational investigations have demonstrated the feasibility of 11C-OMAR or 11C-KR31173 and PET/CT to image and quantify myocardial CB1-R and/or AT1-R expression, respectively. There is an increasing understanding of the mechanisms of activated myocardial CB1-R and/or AT1-R to influence the left-ventricular remodeling process in systolic heart failure in different disease entities. The review summarizes contributions of PET to image myocardial CB1-R and AT1-R expression that may have the potential to serve as a target to tailor preventive medical care in the individual patient.
Collapse
Affiliation(s)
- Ines Valenta
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Washington University School of Medicine, Washington University in St. Louis, 510 S. Kingshighway Boulevard, Campus Box 8223, St. Louis, MO, 63110, USA
| | - Pal Pacher
- Laboratory of Physiological Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Vasken Dilsizian
- Department of Diagnostic Radiology and Nuclear Medicine, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Thomas H Schindler
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Washington University School of Medicine, Washington University in St. Louis, 510 S. Kingshighway Boulevard, Campus Box 8223, St. Louis, MO, 63110, USA.
| |
Collapse
|
16
|
Sloan ME, Grant CW, Gowin JL, Ramchandani VA, Le Foll B. Endocannabinoid signaling in psychiatric disorders: a review of positron emission tomography studies. Acta Pharmacol Sin 2019; 40:342-350. [PMID: 30166624 PMCID: PMC6460371 DOI: 10.1038/s41401-018-0081-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/18/2018] [Indexed: 12/28/2022]
Abstract
Endocannabinoid signaling is implicated in an array of psychopathologies ranging from anxiety to psychosis and addiction. In recent years, radiotracers targeting the endocannabinoid system have been used in positron emission tomography (PET) studies to determine whether individuals with psychiatric disorders display altered endocannabinoid signaling. We comprehensively reviewed PET studies examining differences in endocannabinoid signaling between individuals with psychiatric illness and healthy controls. Published studies evaluated individuals with five psychiatric disorders: cannabis use disorder, alcohol use disorder, schizophrenia, post-traumatic stress disorder, and eating disorders. Most studies employed radiotracers targeting cannabinoid receptor 1 (CB1). Cannabis users consistently demonstrated decreased CB1 binding compared to controls, with normalization following short periods of abstinence. Findings in those with alcohol use disorder and schizophrenia were less consistent, with some studies demonstrating increased CB1 binding and others demonstrating decreased CB1 binding. Evidence of aberrant CB1 binding was also found in individuals with anorexia nervosa and post-traumatic stress disorder, but limited data have been published to date. Thus, existing evidence suggests that alterations in endocannabinoid signaling are present in a range of psychiatric disorders. Although recent efforts have largely focused on evaluating CB1 binding, the synthesis of new radiotracers targeting enzymes involved in endocannabinoid degradation, such as fatty acid amide hydrolase, will allow for other facets of endocannabinoid signaling to be evaluated in future studies.
Collapse
Affiliation(s)
- Matthew E Sloan
- Section on Human Psychopharmacology, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20814, USA
| | - Caroline W Grant
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20814, USA
| | - Joshua L Gowin
- Section on Human Psychopharmacology, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20814, USA
| | - Vijay A Ramchandani
- Section on Human Psychopharmacology, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20814, USA
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M5S 2S1, Canada.
- Addiction Medicine Service, Centre for Addiction and Mental Health, Toronto, ON, M6J 1H4, Canada.
- Departments of Family and Community Medicine, Pharmacology and Toxicology, Psychiatry, Institute of Medical Science, University of Toronto, Toronto, ON, M5S 2S1, Canada.
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M6J 1H4, Canada.
| |
Collapse
|
17
|
Xu Y, Li Z. Imaging metabotropic glutamate receptor system: Application of positron emission tomography technology in drug development. Med Res Rev 2019; 39:1892-1922. [DOI: 10.1002/med.21566] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Youwen Xu
- Independent Consultant and Contractor, Radiopharmaceutical Development, Validation and Bio-Application; Philadelphia Pennsylvania
| | - Zizhong Li
- Pharmaceutical Research and Development, SOFIE Biosciences; Somerset New Jersey
| |
Collapse
|
18
|
Chye Y, Christensen E, Solowij N, Yücel M. The Endocannabinoid System and Cannabidiol's Promise for the Treatment of Substance Use Disorder. Front Psychiatry 2019; 10:63. [PMID: 30837904 PMCID: PMC6390812 DOI: 10.3389/fpsyt.2019.00063] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/28/2019] [Indexed: 11/16/2022] Open
Abstract
Substance use disorder is characterized by repeated use of a substance, leading to clinically significant distress, making it a serious public health concern. The endocannabinoid system plays an important role in common neurobiological processes underlying substance use disorder, in particular by mediating the rewarding and motivational effects of substances and substance-related cues. In turn, a number of cannabinoid drugs (e.g., rimonabant, nabiximols) have been suggested for potential pharmacological treatment for substance dependence. Recently, cannabidiol (CBD), a non-psychoactive phytocannabinoid found in the cannabis plant, has also been proposed as a potentially effective treatment for the management of substance use disorder. Animal and human studies suggest that these cannabinoids have the potential to reduce craving and relapse in abstinent substance users, by impairing reconsolidation of drug-reward memory, salience of drug cues, and inhibiting the reward-facilitating effect of drugs. Such functions likely arise through the targeting of the endocannabinoid and serotonergic systems, although the exact mechanism is yet to be elucidated. This article seeks to review the role of the endocannabinoid system in substance use disorder and the proposed pharmacological action supporting cannabinoid drugs' therapeutic potential in addictions, with a focus on CBD. Subsequently, this article will evaluate the underlying evidence for CBD as a potential treatment for substance use disorder, across a range of substances including nicotine, alcohol, psychostimulants, opioids, and cannabis. While early research supports CBD's promise, further investigation and validation of CBD's efficacy, across preclinical and clinical trials will be necessary.
Collapse
Affiliation(s)
- Yann Chye
- Brain and Mental Health Research Hub, Monash Institute of Cognitive and Clinical Neurosciences, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Erynn Christensen
- Brain and Mental Health Research Hub, Monash Institute of Cognitive and Clinical Neurosciences, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | - Nadia Solowij
- School of Psychology and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,The Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW, Australia
| | - Murat Yücel
- Brain and Mental Health Research Hub, Monash Institute of Cognitive and Clinical Neurosciences, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Liow JS, Morse CL, Lu S, Frankland M, Tye GL, Zoghbi SS, Gladding RL, Shaik AB, Innis RB, Newman AH, Pike VW. [ O- methyl- 11C] N-(4-(4-(3-Chloro-2-methoxyphenyl)-piperazin-1-yl)butyl)-1 H-indole-2-carboxamide ([ 11C]BAK4-51) Is an Efflux Transporter Substrate and Ineffective for PET Imaging of Brain D₃ Receptors in Rodents and Monkey. Molecules 2018; 23:molecules23112737. [PMID: 30360553 PMCID: PMC6278341 DOI: 10.3390/molecules23112737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 11/16/2022] Open
Abstract
Selective high-affinity antagonists for the dopamine D₃ receptor (D₃R) are sought for treating substance use disorders. Positron emission tomography (PET) with an effective D₃R radioligand could be a useful tool for the development of such therapeutics by elucidating pharmacological specificity and target engagement in vivo. Currently, a D₃R-selective radioligand does not exist. The D₃R ligand, N-(4-(4-(3-chloro-2-methoxyphenyl)piperazin-1-yl)butyl)-1H-indole-2-carboxamide (BAK4-51, 1), has attractive properties for PET radioligand development, including full antagonist activity, very high D₃R affinity, D₃R selectivity, and moderate lipophilicity. We labeled 1 with the positron-emitter carbon-11 (t1/2 = 20.4 min) in the methoxy group for evaluation as a radioligand in animals with PET. However, [11C]1 was found to be an avid substrate for brain efflux transporters and lacked D₃R-specific signal in rodent and monkey brain in vivo.
Collapse
Affiliation(s)
- Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Shuiyu Lu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Michael Frankland
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - George L Tye
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Robert L Gladding
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Anver B Shaik
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Amy H Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Laurikainen H, Tuominen L, Tikka M, Merisaari H, Armio RL, Sormunen E, Borgan F, Veronese M, Howes O, Haaparanta-Solin M, Solin O, Hietala J. Sex difference in brain CB1 receptor availability in man. Neuroimage 2018; 184:834-842. [PMID: 30296558 DOI: 10.1016/j.neuroimage.2018.10.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 09/20/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022] Open
Abstract
The endocannabinoid system (ECS) has a widespread neuromodulatory function in the central nervous system and is involved in important aspects of brain function including brain development, cortical rhythms, plasticity, reward, and stress sensitivity. Many of these effects are mediated via the cannabinoid CB1 receptor (CB1R) subtype. Animal studies convincingly show an interaction between the ECS and sex hormones, as well as a sex difference of higher brain CB1R in males. Human in vivo studies of sex difference have yielded discrepant findings. Gender differences in CB1R availability were investigated in vivo in 11 male and 11 female healthy volunteers using a specific CB1R tracer [18F]FMPEP-d2 and positron emission tomography (PET). Regional [18F]FMPEP-d2 distribution volume was used as a proxy for CB1R availability. In addition, we explored whether CB1R availability is linked to neuropsychological functioning. Relative to females, CB1R availability was on average 41% higher in males (p = 0.002) with a regionally specific effect larger in the posterior cingulate and retrosplenial cortices (p = 0.001). Inter-subject variability in CB1R availability was similar in both groups. Voxel-based analyses revealed an inverse association between CB1R availability and visuospatial working memory task performance in both groups (p < 0.001). A CB1R sex difference with a large effect size was observed and should be considered in the design of CB1R-related studies on neuropsychiatric disorders. The behavioural correlates and clinical significance of this difference remain to be further elucidated, but our studies suggest an association between CB1R availability and working memory.
Collapse
Affiliation(s)
- Heikki Laurikainen
- Turku PET Centre, Turku University Hospital, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Finland
| | - Lauri Tuominen
- Turku PET Centre, Turku University Hospital, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Finland; Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA, USA
| | - Maria Tikka
- Department of Psychiatry, University of Turku and Turku University Hospital, Finland
| | | | - Reetta-Liina Armio
- Turku PET Centre, Turku University Hospital, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Finland
| | - Elina Sormunen
- Turku PET Centre, Turku University Hospital, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Finland
| | - Faith Borgan
- Psychosis Studies Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Oliver Howes
- Psychosis Studies Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | | | - Olof Solin
- Turku PET Centre, Turku University Hospital, Finland
| | - Jarmo Hietala
- Turku PET Centre, Turku University Hospital, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Finland.
| | | |
Collapse
|
21
|
Frank E, Maier D, Pajula J, Suvitaival T, Borgan F, Butz-Ostendorf M, Fischer A, Hietala J, Howes O, Hyötyläinen T, Janssen J, Laurikainen H, Moreno C, Suvisaari J, Van Gils M, Orešič M. Platform for systems medicine research and diagnostic applications in psychotic disorders-The METSY project. Eur Psychiatry 2018; 50:40-46. [PMID: 29361398 DOI: 10.1016/j.eurpsy.2017.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022] Open
Abstract
Psychotic disorders are associated with metabolic abnormalities including alterations in glucose and lipid metabolism. A major challenge in the treatment of psychosis is to identify patients with vulnerable metabolic profiles who may be at risk of developing cardiometabolic co-morbidities. It is established that both central and peripheral metabolic organs use lipids to control energy balance and regulate peripheral insulin sensitivity. The endocannabinoid system, implicated in the regulation of glucose and lipid metabolism, has been shown to be dysregulated in psychosis. It is currently unclear how these endocannabinoid abnormalities relate to metabolic changes in psychosis. Here we review recent research in the field of metabolic co-morbidities in psychotic disorders as well as the methods to study them and potential links to the endocannabinoid system. We also describe the bioinformatics platforms developed in the EU project METSY for the investigations of the biological etiology in patients at risk of psychosis and in first episode psychosis patients. The METSY project was established with the aim to identify and evaluate multi-modal peripheral and neuroimaging markers that may be able to predict the onset and prognosis of psychiatric and metabolic symptoms in patients at risk of developing psychosis and first episode psychosis patients. Given the intrinsic complexity and widespread role of lipid metabolism, a systems biology approach which combines molecular, structural and functional neuroimaging methods with detailed metabolic characterisation and multi-variate network analysis is essential in order to identify how lipid dysregulation may contribute to psychotic disorders. A decision support system, integrating clinical, neuropsychological and neuroimaging data, was also developed in order to aid clinical decision making in psychosis. Knowledge of common and specific mechanisms may aid the etiopathogenic understanding of psychotic and metabolic disorders, facilitate early disease detection, aid treatment selection and elucidate new targets for pharmacological treatments.
Collapse
Affiliation(s)
| | | | - Juha Pajula
- VTT Technical Research Centre of Finland Ltd., FI-33720 Tampere, Finland
| | | | - Faith Borgan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London WC2R 2LS, UK; Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London W12 0HS, UK
| | | | | | - Jarmo Hietala
- Department of Psychiatry, University of Turku, FI-20520 Turku, Finland; Turku PET Centre, Turku University Hospital, FI-20521 Turku, Finland
| | - Oliver Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London WC2R 2LS, UK; Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London W12 0HS, UK
| | | | - Joost Janssen
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Heikki Laurikainen
- Department of Psychiatry, University of Turku, FI-20520 Turku, Finland; Turku PET Centre, Turku University Hospital, FI-20521 Turku, Finland
| | - Carmen Moreno
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Jaana Suvisaari
- National Institute for Health and Welfare (THL), FI-00300 Helsinki, Finland
| | - Mark Van Gils
- VTT Technical Research Centre of Finland Ltd., FI-33720 Tampere, Finland
| | - Matej Orešič
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland; School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden.
| |
Collapse
|
22
|
Valenta I, Varga ZV, Valentine H, Cinar R, Horti A, Mathews WB, Dannals RF, Steele K, Kunos G, Wahl RL, Pomper MG, Wong DF, Pacher P, Schindler TH. Feasibility Evaluation of Myocardial Cannabinoid Type 1 Receptor Imaging in Obesity: A Translational Approach. JACC Cardiovasc Imaging 2018; 11:320-332. [PMID: 29413441 PMCID: PMC6178217 DOI: 10.1016/j.jcmg.2017.11.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 11/15/2017] [Accepted: 11/30/2017] [Indexed: 11/24/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the feasibility of targeted imaging of myocardial cannabinoid type 1 receptor (CB1-R) and its potential up-regulation in obese mice with translation to humans using [11C]-OMAR and positron emission tomography (PET)/computed tomography (CT). BACKGROUND Activation of myocardial CB1-R by endocannabinoids has been implicated in cardiac dysfunction in diabetic mice. Obesity may lead to an up-regulation of myocardial CB1-R, potentially providing a mechanistic link between obesity and the initiation and/or progression of cardiomyopathy. METHODS Binding specificity of [11C]-OMAR to CB1-R was investigated by blocking studies with rimonabant in mice. The heart was harvested from each mouse, and its radioactivity was determined by γ-counter. Furthermore, [11C]-OMAR dynamic micro-PET/CT was carried out in obese and normal-weight mice. Ex vivo validation was performed by droplet digital polymerase chain reaction (absolute quantification) and RNAscope Technology (an in situ ribonucleic acid analysis platform). Subsequently, myocardial CB1-R expression was probed noninvasively with intravenous injection of CB1-R ligand [11C]-OMAR and PET/CT in humans with advanced obesity and normal-weight human control subjects, respectively. RESULTS Rimonabant significantly blocked OMAR uptake in the heart muscle compared with vehicle, signifying specific binding of OMAR to the CB1-R in the myocardium. The myocardial OMAR retention quantified by micro-PET/CT in mice was significantly higher in obese compared with normal-weight mice. Absolute quantification of CB1-R gene expression with droplet digital polymerase chain reaction and in situ hybridization confirmed CB1-R up-regulation in all major myocardial cell types (e.g., cardiomyocytes, endothelium, vascular smooth muscle cells, and fibroblasts) of obese mice. Obese mice also had elevated myocardial levels of endocannabinoids anandamide and 2-arachidonoylglycerol compared with lean mice. Translation to humans revealed higher myocardial OMAR retention in advanced obesity compared with normal-weight subjects. CONCLUSIONS Noninvasive imaging of cardiac CB1-R expression in obesity is feasible applying [11C]-OMAR and PET/CT. These results may provide a rationale for further clinical testing of CB1-R-targeted molecular imaging in cardiometabolic diseases.
Collapse
Affiliation(s)
- Ines Valenta
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zoltan V Varga
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Heather Valentine
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Resat Cinar
- Laboratory of Physiological Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Andrew Horti
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William B Mathews
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F Dannals
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kimberley Steele
- Department of Surgery, Bariatric Center at Bayview, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George Kunos
- Laboratory of Physiological Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Richard L Wahl
- Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, Missouri
| | - Martin G Pomper
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dean F Wong
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland.
| | - Thomas H Schindler
- Department of Radiology, Division of Nuclear Medicine, Nuclear Cardiovascular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
23
|
Fisher MJ, McMurray L, Lu S, Morse CL, Liow JS, Zoghbi SS, Kowalski A, Tye GL, Innis RB, Aigbirhio FI, Pike VW. [Carboxyl- 11 C]Labelling of Four High-Affinity cPLA2α Inhibitors and Their Evaluation as Radioligands in Mice by Positron Emission Tomography. ChemMedChem 2018; 13:138-146. [PMID: 29232493 DOI: 10.1002/cmdc.201700697] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/11/2017] [Indexed: 01/23/2023]
Abstract
Cytosolic phospholipase A2α (cPLA2α) may play a critical role in neuropsychiatric and neurodegenerative disorders associated with oxidative stress and neuroinflammation. An effective PET radioligand for imaging cPLA2α in living brain might prove useful for biomedical research, especially on neuroinflammation. We selected four high-affinity (IC50 2.1-12 nm) indole-5-carboxylic acid-based inhibitors of cPLA2α, namely 3-isobutyryl-1-(2-oxo-3-(4-phenoxyphenoxy)propyl)-1H-indole-5-carboxylic acid (1); 3-acetyl-1-(2-oxo-3-(4-(4-(trifluoromethyl)phenoxy)phenoxy)propyl)-1H-indole-5-carboxylic acid (2); 3-(3-methyl-1,2,4-oxadiazol-5-yl)-1-(2-oxo-3-(4-phenoxyphenoxy)propyl)-1H-indole-5-carboxylic acid (3); and 3-(3-methyl-1,2,4-oxadiazol-5-yl)-1-(3-(4-octylphenoxy)-2-oxopropyl)-1H-indole-5-carboxylic acid (4), for labelling in carboxyl position with carbon-11 (t1/2 =20.4 min) to provide candidate PET radioligands for imaging brain cPLA2α. Compounds [11 C]1-4 were obtained for intravenous injection in adequate overall yields (1.1-5.5 %) from cyclotron-produced [11 C]carbon dioxide and with moderate molar activities (70-141 GBq μmol-1 ) through the use of Pd0 -mediated [11 C]carbon monoxide insertion on iodo precursors. Measured logD7.4 values were within a narrow moderate range (1.9-2.4). After intravenous injection of [11 C]1-4 in mice, radioactivity uptakes in brain peaked at low values (≤0.8 SUV) and decreased by about 90 % over 15 min. Pretreatments of the mice with high doses of the corresponding non-radioactive ligands did not alter brain time-activity curves. Brain uptakes of radioactivity after administration of [11 C]1 to wild-type and P-gp/BCRP dual knock-out mice were similar (peak 0.4 vs. 0.5 SUV), indicating that [11 C]1 and others in this structural class, are not substrates for efflux transporters.
Collapse
Affiliation(s)
- Martin J Fisher
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Lindsay McMurray
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Shuiyu Lu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| | - Aneta Kowalski
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| | - George L Tye
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| | - Franklin I Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Miederer I, Buchholz HG, Kronfeld A, Maus S, Weyer-Elberich V, Mildenberger P, Lutz B, Schreckenberger M. Pharmacokinetics of the cannabinoid receptor ligand [18
F]MK-9470 in the rat brain - Evaluation of models using microPET. Med Phys 2018; 45:725-734. [DOI: 10.1002/mp.12732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/24/2017] [Accepted: 11/30/2017] [Indexed: 12/19/2022] Open
Affiliation(s)
- Isabelle Miederer
- Department of Nuclear Medicine; University Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstraße 1 55131 Mainz Germany
| | - Hans-Georg Buchholz
- Department of Nuclear Medicine; University Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstraße 1 55131 Mainz Germany
| | - Andrea Kronfeld
- Institute of Microscopic Anatomy and Neurobiology; University Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstraße 1 55131 Mainz Germany
| | - Stephan Maus
- Department of Nuclear Medicine; University Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstraße 1 55131 Mainz Germany
| | - Veronika Weyer-Elberich
- Institute of Medical Biostatistics, Epidemiology and Informatics; University Medical Center of the Johannes Gutenberg University Mainz; Obere Zahlbacher Straße 69 55131 Mainz Germany
| | - Philipp Mildenberger
- Institute of Medical Biostatistics, Epidemiology and Informatics; University Medical Center of the Johannes Gutenberg University Mainz; Obere Zahlbacher Straße 69 55131 Mainz Germany
| | - Beat Lutz
- Institute of Physiological Chemistry; University Medical Center of the Johannes Gutenberg University Mainz; Duesbergweg 6 55128 Mainz Germany
| | - Mathias Schreckenberger
- Department of Nuclear Medicine; University Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstraße 1 55131 Mainz Germany
| |
Collapse
|
25
|
Cilia R. Molecular Imaging of the Cannabinoid System in Idiopathic Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 141:305-345. [DOI: 10.1016/bs.irn.2018.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
|
26
|
Schifani C, Hafizi S, Da Silva T, Watts JJ, Khan MS, Mizrahi R. Using molecular imaging to understand early schizophrenia-related psychosis neurochemistry: a review of human studies. Int Rev Psychiatry 2017; 29:555-566. [PMID: 29219634 PMCID: PMC8011813 DOI: 10.1080/09540261.2017.1396205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Schizophrenia is a chronic psychiatric disorder generally preceded by a so-called prodromal phase, which is characterized by attenuated psychotic symptoms. Advances in clinical research have enabled prospective identification of those individuals who are at clinical high risk (CHR) for psychosis, with the power to predict psychosis onset within the near future. Changes in several brain neurochemical systems and molecular mechanisms are implicated in the pathophysiology of schizophrenia and the psychosis spectrum, including the dopaminergic, γ-aminobutyric acid (GABA)-ergic, glutamatergic, endocannabinoid, and immunologic (i.e. glial activation) system and other promising future directions such as synaptic density, which are possible to quantify in vivo using positron emission tomography (PET). This paper aims to review in vivo PET studies in the mentioned systems in the early course of psychosis (i.e. CHR and first-episode psychosis (FEP)). The results of reviewed studies are promising; however, the current understanding of the underlying pathology of psychosis is still limited. Importantly, promising efforts involve the development of novel PET radiotracers targeting systems with growing interest in schizophrenia, like the nociceptive system and synaptic density.
Collapse
Affiliation(s)
- Christin Schifani
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Sina Hafizi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tania Da Silva
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jeremy Joseph Watts
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - M. Saad Khan
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Romina Mizrahi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
27
|
Bhattacharyya S, Egerton A, Kim E, Rosso L, Riano Barros D, Hammers A, Brammer M, Turkheimer FE, Howes OD, McGuire P. Acute induction of anxiety in humans by delta-9-tetrahydrocannabinol related to amygdalar cannabinoid-1 (CB1) receptors. Sci Rep 2017; 7:15025. [PMID: 29101333 PMCID: PMC5670208 DOI: 10.1038/s41598-017-14203-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022] Open
Abstract
Use of Cannabis, the most widely used illicit drug worldwide, is associated with acute anxiety, and anxiety disorders following regular use. The precise neural and receptor basis of these effects have not been tested in man. Employing a combination of functional MRI (fMRI) and positron emission tomography (PET), we investigated whether the effects of delta-9-tetrahydrocannabinol (delta-9-THC), the main psychoactive ingredient of cannabis, on anxiety and on amygdala response while processing fearful stimuli were related to local availability of its main central molecular target, cannabinoid-1 (CB1) receptors in man. Fourteen healthy males were studied with fMRI twice, one month apart, following an oral dose of either delta-9-THC (10 mg) or placebo, while they performed a fear-processing task. Baseline availability of the CB1 receptor was studied using PET with [11C]MePPEP, a CB1 inverse agonist radioligand. Relative to the placebo condition, delta-9-THC induced anxiety and modulated right amygdala activation while processing fear. Both these effects were positively correlated with CB1 receptor availability in the right amygdala. These results suggest that the acute effects of cannabis on anxiety in males are mediated by the modulation of amygdalar function by delta-9-THC and the extent of these effects are related to local availability of CB1 receptors.
Collapse
Affiliation(s)
- Sagnik Bhattacharyya
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK.
| | - Alice Egerton
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Euitae Kim
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Lula Rosso
- Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | | | - Alexander Hammers
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, 4th floor Lambeth Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Michael Brammer
- Department of Neuroimaging, Centre for Neuroimaging Sciences, PO Box 089, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Federico E Turkheimer
- Department of Neuroimaging, Centre for Neuroimaging Sciences, PO Box 089, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Oliver D Howes
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
- Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| |
Collapse
|
28
|
Abstract
Purpose of Review To review the current status of positron emission tomography (PET) molecular imaging research of levodopa-induced dyskinesias (LIDs) in Parkinson’s disease (PD). Recent Findings Recent PET studies have provided robust evidence that LIDs in PD are associated with elevated and fluctuating striatal dopamine synaptic levels, which is a consequence of the imbalance between dopaminergic and serotonergic terminals, with the latter playing a key role in mishandling presynaptic dopamine release. Long-term exposure to levodopa is no longer believed to solely induce LIDs, as studies have highlighted that PD patients who go on to develop LIDs exhibit elevated putaminal dopamine release before the initiation of levodopa treatment, suggesting the involvement of other mechanisms, including altered neuronal firing and abnormal levels of phosphodiesterase 10A. Summary Dopaminergic, serotonergic, glutamatergic, adenosinergic and opioid systems and phosphodiesterase 10A levels have been shown to be implicated in the development of LIDs in PD. However, no system may be considered sufficient on its own for the development of LIDs, and the mechanisms underlying LIDs in PD may have a multisystem origin. In line with this notion, future studies should use multimodal PET molecular imaging in the same individuals to shed further light on the different mechanisms underlying the development of LIDs in PD.
Collapse
|
29
|
Wilson H, De Micco R, Niccolini F, Politis M. Molecular Imaging Markers to Track Huntington's Disease Pathology. Front Neurol 2017; 8:11. [PMID: 28194132 PMCID: PMC5278260 DOI: 10.3389/fneur.2017.00011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/09/2017] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is a progressive, monogenic dominant neurodegenerative disorder caused by repeat expansion mutation in the huntingtin gene. The accumulation of mutant huntingtin protein, forming intranuclear inclusions, subsequently leads to degeneration of medium spiny neurons in the striatum and cortical areas. Genetic testing can identify HD gene carriers before individuals develop overt cognitive, psychiatric, and chorea symptoms. Thus, HD gene carriers can be studied in premanifest stages to understand and track the evolution of HD pathology. While advances have been made, the precise pathophysiological mechanisms underlying HD are unclear. Magnetic resonance imaging (MRI) and positron emission tomography (PET) have been employed to understand HD pathology in presymptomatic and symptomatic disease stages. PET imaging uses radioactive tracers to detect specific changes, at a molecular level, which could be used as markers of HD progression and to monitor response to therapeutic treatments for HD gene expansion carriers (HDGECs). This review focuses on available PET techniques, employed in cross-sectional and longitudinal human studies, as biomarkers for HD, and highlights future potential PET targets. PET studies have assessed changes in postsynaptic dopaminergic receptors, brain metabolism, microglial activation, and recently phosphodiesterase 10A (PDE10A) as markers to track HD progression. Alterations in PDE10A expression are the earliest biochemical change identified in HD gene carriers up to 43 years before predicted symptomatic onset. Thus, PDE10A expression could be a promising marker to track HD progression from early premanifest disease stages. Other PET targets which have been less well investigated as biomarkers include cannabinoid, adenosine, and GABA receptors. Future longitudinal studies are required to fully validate these PET biomarkers for use to track disease progression from far-onset premanifest to manifest HD stages. PET imaging is a crucial neuroimaging tool, with the potential to detect early changes and validate sensitivity of biomarkers for tracking HD pathology. Moreover, continued development of novel PET tracers provides exciting opportunities to investigate new molecular targets, such as histamine and serotonin receptors, to further understand the mechanisms underlying HD pathology.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| | - Rosa De Micco
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| | - Flavia Niccolini
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| |
Collapse
|
30
|
Takano A, Varrone A, Gulyás B, Salvadori P, Gee A, Windhorst A, Vercouillie J, Bormans G, Lammertsma AA, Halldin C. Guidelines to PET measurements of the target occupancy in the brain for drug development. Eur J Nucl Med Mol Imaging 2016; 43:2255-2262. [PMID: 27514528 PMCID: PMC5047931 DOI: 10.1007/s00259-016-3476-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 11/30/2022]
Abstract
This guideline summarizes the current view of the European Association of Nuclear Medicine Drug Development Committee. The purpose of this guideline is to guarantee a high standard of PET studies that are aimed at measuring target occupancy in the brain within the framework of development programs of drugs that act within the central nervous system (CNS drugs). This guideline is intended to present information specifically adapted to European practice. The information provided should be applied within the context of local conditions and regulations.
Collapse
Affiliation(s)
- Akihiro Takano
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden.
| | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Balázs Gulyás
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | | | - Antony Gee
- Department of Chemistry and Biology, Division of Imaging Sciences and Biomedical Engineering, Kings College London, London, UK
| | - Albert Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Guy Bormans
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Tonietto M, Rizzo G, Veronese M, Bertoldo A. Modelling arterial input functions in positron emission tomography dynamic studies. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2016; 2015:2247-50. [PMID: 26736739 DOI: 10.1109/embc.2015.7318839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The quantification of dynamic positron emission tomography (PET) images often requires the invasive measures of the arterial plasma tracer concentration to be used as arterial input function (AIF). In several situations, a mathematical model is fit to the hematic data to obtain a continuous and noise-free description of the AIF. In common practice, the tri-exponential and Feng's models are generally adopted. Despite their general applicability, often these approximations of blood tracer activity do not properly describe the complex behavior of the AIF (e.g. different clearance rates of the tracers) as well as they do not account for the length of the radiotracer injection. Here we propose two models able to include the injection duration as additional information in the AIF modeling and we compare their performances in eight different datasets acquired from different PET facilities.
Collapse
|
32
|
Lisdahl KM, Tamm L, Epstein JN, Jernigan T, Molina BSG, Hinshaw SP, Swanson JM, Newman E, Kelly C, Bjork JM. The impact of ADHD persistence, recent cannabis use, and age of regular cannabis use onset on subcortical volume and cortical thickness in young adults. Drug Alcohol Depend 2016; 161:135-46. [PMID: 26897585 PMCID: PMC5289096 DOI: 10.1016/j.drugalcdep.2016.01.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/23/2015] [Accepted: 01/26/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Both Attention Deficit Hyperactivity Disorder (ADHD) and chronic cannabis (CAN) use have been associated with brain structural abnormalities, although little is known about the effects of both in young adults. METHODS Participants included: those with a childhood diagnosis of ADHD who were CAN users (ADHD_CAN; n=37) and non-users (NU) (ADHD_NU; n=44) and a local normative comparison group (LNCG) who did (LNCG_CAN; n=18) and did not (LNCG_NU; n=21) use CAN regularly. Multiple regressions and MANCOVAs were used to examine the independent and interactive effects of a childhood ADHD diagnosis and CAN group status and age of onset (CUO) on subcortical volumes and cortical thickness. RESULTS After controlling for age, gender, total brain volume, nicotine use, and past-year binge drinking, childhood ADHD diagnosis did not predict brain structure; however, persistence of ADHD was associated with smaller left precentral/postcentral cortical thickness. Compared to all non-users, CAN users had decreased cortical thickness in right hemisphere superior frontal sulcus, anterior cingulate, and isthmus of cingulate gyrus regions and left hemisphere superior frontal sulcus and precentral gyrus regions. Early cannabis use age of onset (CUO) in those with ADHD predicted greater right hemisphere superior frontal and postcentral cortical thickness. DISCUSSION Young adults with persistent ADHD demonstrated brain structure abnormalities in regions underlying motor control, working memory and inhibitory control. Further, CAN use was linked with abnormal brain structure in regions with high concentrations of cannabinoid receptors. Additional large-scale longitudinal studies are needed to clarify how substance use impacts neurodevelopment in youth with and without ADHD.
Collapse
Affiliation(s)
- Krista M Lisdahl
- University of Wisconsin-Milwaukee, Psychology Department, 2441 E. Hartford Ave, Milwaukee, WI 53211, United States.
| | - Leanne Tamm
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML10006, Cincinnati, OH 45229, United States
| | - Jeffery N Epstein
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML10006, Cincinnati, OH 45229, United States
| | - Terry Jernigan
- University of California, San Diego, 9500 Gilman Drive #0115, La Jolla, CA 92093, United States
| | - Brooke S G Molina
- University of Pittsburgh School of Medicine, Department of Psychology, 3811 O'Hara St., Pittsburgh, PA 15213, United States
| | - Stephen P Hinshaw
- University of California-Berkeley, Department of Psychology, Tolman Hall, Berkeley, CA 94720-1650, United States
| | - James M Swanson
- University of California, Irvine, 19722 MacArthur Boulevard, Irvine, CA 92612, United States
| | - Erik Newman
- University of California, San Diego, 9500 Gilman Drive #0115, La Jolla, CA 92093, United States
| | - Clare Kelly
- The Child Center at New York University, Langone Medical Center, New York, NY 10016, United States
| | - James M Bjork
- Department of Psychiatry, Virginia Commonwealth University, United States
| |
Collapse
|
33
|
Shoup TM, Bonab AA, Wilson AA, Vasdev N. Synthesis and preclinical evaluation of [¹⁸F]FCHC for neuroimaging of fatty acid amide hydrolase. Mol Imaging Biol 2015; 17:257-63. [PMID: 25273322 DOI: 10.1007/s11307-014-0789-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE Fatty acid amide hydrolase (FAAH), a catabolic enzyme which regulates lipid transmitters in the endocannabinoid system, is an avidly sought therapeutic and positron emission tomography (PET) imaging target for studies involving addiction and neurological disorders. We report the synthesis of a new fluorine-18-labeled FAAH inhibitor, trans-3-(4, 5-dihydrooxazol-2-yl)phenyl-4-[(18)F]fluorocyclohexylcarbamate ([(18)F]FCHC), and its evaluation in rat brain. PROCEDURES The synthesis of [(18)F]FCHC was conducted via a 3-step, 1-pot reaction, resulting in uncorrected radiochemical yields between 10 and 20% (n = 5) relative to [(18)F]fluoride, with specific activities of >5 Ci/μmol at the end of the synthesis. The radiosynthesis was seamlessly automated using a commercial radiofluorination apparatus. Ex vivo biodistribution and preliminary PET imaging studies were carried out in male Sprague-Dawley rats. RESULTS Rat brain biodistribution at 2 min post-injection showed a standard uptake value of 4.6 ± 0.1 in the cortex, which increased to 7.8 ± 0.1 at 40 min. Pretreatment with the selective FAAH inhibitor URB597 reduced uptake of radioactivity in all brain regions by >90%, with 98 % blockade in the FAAH-rich cortex. PET imaging was consistent with biodistribution studies. CONCLUSIONS [(18)F]FCHC appears to be a highly sensitive (18)F-labeled radiotracer for imaging FAAH in the central nervous system, and these results warrant further imaging in nonhuman primates.
Collapse
Affiliation(s)
- Timothy M Shoup
- Division of Nuclear Medicine and Molecular Imaging, Center for Advanced Medical Imaging Sciences, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
34
|
Shollenbarger SG, Price J, Wieser J, Lisdahl K. Impact of cannabis use on prefrontal and parietal cortex gyrification and surface area in adolescents and emerging adults. Dev Cogn Neurosci 2015; 16:46-53. [PMID: 26233614 PMCID: PMC5289075 DOI: 10.1016/j.dcn.2015.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Regions undergoing maturation with CB1 receptors may be at increased risk for cannabis-induced alterations. Here, we examine the relationships between cannabis use and prefrontal (PFC) and inferior parietal gyrification and surface area (SA) in youth. METHODS Participants included 33 cannabis users and 35 controls (ages 18-25). Exclusions included co-morbid psychiatric/neurologic disorders and heavy other drug use. Multiple regressions and Pearson r correlations examined the effects of cannabis use on gyrification, SA and cognition. RESULTS Cannabis use was associated with decreased gyrification in: ventral-medial PFC (RH: [FDR corrected p=.02], LH: [FDR corrected p=.02]); medial PFC (RH: [FDR corrected p=.02], LH: [FDR corrected p=.02]); and frontal poles (RH: [FDR corrected p=.02], LH: [FDR corrected p=.02]). No differences were observed in bilateral hemispheres, PFC, dorsolateral, ventrolateral, or inferior parietal ROIs. Cannabis use was associated with marginally decreased SA in left: medial PFC [FDR corrected p=.09], and ventral lateral PFC: [FDR corrected p=.09]. In cannabis users, increased gyrification was associated with improved working-memory performance in right medial (p=.003), ventral-medial (p=.03), and frontal pole ROIs (p=.007). CONCLUSIONS Cannabis use was associated with reduced gyrification in PFC regions implicated in self-referential thought and social cognition. Results suggest that these gyrification characteristics may have cognitive implications.
Collapse
Affiliation(s)
- Skyler G Shollenbarger
- Department of Psychology, University of Wisconsin-Milwaukee, Garland Hall Rm 224, 2441 East Hartford Ave, Milwaukee, WI 53211, United States.
| | - Jenessa Price
- McLean Hospital-Harvard Medical School, 115 Mill St., Belmont, MA 02478, United States.
| | - Jon Wieser
- Department of Psychology, University of Wisconsin-Milwaukee, Garland Hall Rm 224, 2441 East Hartford Ave, Milwaukee, WI 53211, United States.
| | - Krista Lisdahl
- Department of Psychology, University of Wisconsin-Milwaukee, Garland Hall Rm 224, 2441 East Hartford Ave, Milwaukee, WI 53211, United States.
| |
Collapse
|
35
|
Gunn RN, Slifstein M, Searle GE, Price JC. Quantitative imaging of protein targets in the human brain with PET. Phys Med Biol 2015; 60:R363-411. [DOI: 10.1088/0031-9155/60/22/r363] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Improved models for plasma radiometabolite correction and their impact on kinetic quantification in PET studies. J Cereb Blood Flow Metab 2015; 35:1462-9. [PMID: 25873424 PMCID: PMC4640335 DOI: 10.1038/jcbfm.2015.61] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 11/08/2022]
Abstract
The quantification of dynamic positron emission tomography studies performed with arterial sampling usually requires correcting the input function for the presence of radiometabolites by using a model of the plasma parent fraction (PPf). Here, we show how to include the duration of radioligand injection in the PPf model formulations to achieve a more physiologic description of the plasma measurements. This formulation (here called convoluted model) was tested on simulated data and on three datasets with different parent kinetics: [(11)C]NOP-1A, [(11)C]MePPEP, and [(11)C](R)-rolipram. Results showed that convoluted PPf models better described the fraction of unchanged parent in the plasma compared with standard models for all three datasets (weighted residuals sum of squares up to 25% lower). When considering the effect on tissue quantification, the overall impact on the total volume of distribution (VT) was low. However, the impact was significant and radioligand-dependent on the binding potential (BP) and the microparameters (K1, k2, k3, and k4). Simulated data confirmed that quantification is sensitive to different degrees to PPf model misspecification. Including the injection duration allows obtaining a more accurate correction of the input function for the presence of radiometabolites and this yields a more reliable quantification of the tissue parameters.
Collapse
|
37
|
Imaging the cannabinoid CB1 receptor in humans with [11C]OMAR: assessment of kinetic analysis methods, test-retest reproducibility, and gender differences. J Cereb Blood Flow Metab 2015; 35:1313-22. [PMID: 25833345 PMCID: PMC4528005 DOI: 10.1038/jcbfm.2015.46] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 02/05/2015] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
The Radiotracer [(11)C]OMAR was developed for positron emission tomography (PET) imaging of cannabinoid type-1 receptors (CB1R). The objectives of the present study were to evaluate kinetic analysis methods, determine test-retest reliability, and assess gender differences in receptor availability. Dynamic PET data were acquired in 10 human subjects, and analyzed with one-tissue (1T) and two-tissue (2T) compartment models and by the Logan and multilinear analysis (MA1) methods to estimate regional volume of distribution (VT). The 2T model inclusive of a vascular component (2TV) and MA1 were the preferred techniques. Test-retest reliability of VT was good (mean absolute deviation ~9%; intraclass correlation coefficient ~0.7). Tracer parent fraction in plasma was lower in women (P<0.0001). Cerebral uptake normalized by body weight and injected dose was higher in men by 17% (P<0.0001), but VT was significantly greater in women by 23% (P<0.0001). These findings show that [(11)C]OMAR binding can be reliably quantified by the 2T model or MA1 method and demonstrate the utility of this tracer for in vivo imaging of CB1R. In addition, results from the present study indicate that gender difference in receptor binding should be taken into consideration when [(11)C]OMAR is used to quantify CB1R availability in neuropsychiatric disorders.
Collapse
|
38
|
Shollenbarger SG, Price J, Wieser J, Lisdahl K. Poorer frontolimbic white matter integrity is associated with chronic cannabis use, FAAH genotype, and increased depressive and apathy symptoms in adolescents and young adults. NEUROIMAGE-CLINICAL 2015; 8:117-25. [PMID: 26106535 PMCID: PMC4473294 DOI: 10.1016/j.nicl.2015.03.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 03/17/2015] [Accepted: 03/28/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND The heaviest period of cannabis use coincides with ongoing white matter (WM) maturation. Further, cannabis-related changes may be moderated by FAAH genotype (rs324420). We examined the association between cannabis use and FAAH genotype on frontolimbic WM integrity in adolescents and emerging adults. We then tested whether observed WM abnormalities were linked with depressive or apathy symptoms. METHODS Participants included 37 cannabis users and 37 healthy controls (33 female; ages 18-25). Multiple regressions examined the independent and interactive effects of variables on WM integrity. RESULTS Regular cannabis users demonstrated reduced WM integrity in the bilateral uncinate fasciculus (UNC) (MD, right: p = .009 and left: p = .009; FA, right: p = .04 and left: p = .03) and forceps minor (fMinor) (MD, p = .03) compared to healthy controls. Marginally reduced WM integrity in the cannabis users was found in the left anterior thalamic radiation (ATR) (FA, p = .08). Cannabis group ∗ FAAH genotype interaction predicted WM integrity in bilateral ATR (FA, right: p = .05 and left: p = .001) and fMinor (FA, p = .02). In cannabis users, poorer WM integrity was correlated with increased symptoms of depression and apathy in bilateral ATR and UNC. CONCLUSIONS Consistent with prior findings, cannabis use was associated with reduced frontolimbic WM integrity. WM integrity was also moderated by FAAH genotype, in that cannabis-using FAAH C/C carriers and A carrying controls had reduced WM integrity compared to control C/C carriers. Observed frontolimbic white matter abnormalities were linked with increased depressive and apathy symptoms in the cannabis users.
Collapse
Affiliation(s)
- Skyler G Shollenbarger
- Department of Psychology, University of Wisconsin-Milwaukee, Garland Hall Rm 224, 2441 East Hartford Ave, Milwaukee, WI 53211, USA
| | - Jenessa Price
- McLean Hospital, Harvard Medical School, 115 Mill St., Belmont, MA 02478, USA
| | - Jon Wieser
- Department of Psychology, University of Wisconsin-Milwaukee, Garland Hall Rm 224, 2441 East Hartford Ave, Milwaukee, WI 53211, USA
| | - Krista Lisdahl
- Department of Psychology, University of Wisconsin-Milwaukee, Garland Hall Rm 224, 2441 East Hartford Ave, Milwaukee, WI 53211, USA
| |
Collapse
|
39
|
Hjorth S, Karlsson C, Jucaite A, Varnäs K, Wählby Hamrén U, Johnström P, Gulyás B, Donohue SR, Pike VW, Halldin C, Farde L. A PET study comparing receptor occupancy by five selective cannabinoid 1 receptor antagonists in non-human primates. Neuropharmacology 2015; 101:519-30. [PMID: 25791528 DOI: 10.1016/j.neuropharm.2015.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/05/2015] [Accepted: 03/07/2015] [Indexed: 01/14/2023]
Abstract
There is a medical need for safe and efficacious anti-obesity drugs with acceptable side effect profiles. To mitigate the challenge posed by translating target interaction across species and balancing beneficial vs. adverse effects, a positron emission tomography (PET) approach could help guide clinical dose optimization. Thus, as part of a compound differentiation effort, three novel selective CB1 receptor (CB1R) antagonists, developed by AstraZeneca (AZ) for the treatment of obesity, were compared with two clinically tested reference compounds, rimonabant and taranabant, with regard to receptor occupancy relative to dose and exposure. A total of 42 PET measurements were performed in 6 non-human primates using the novel CB1R antagonist radioligand [(11)C]SD5024. The AZ CB1R antagonists bound in a saturable manner to brain CB1R with in vivo affinities similar to that of rimonabant and taranabant, compounds with proven weight loss efficacy in clinical trials. Interestingly, it was found that exposures corresponding to those needed for optimal clinical efficacy of rimonabant and taranabant resulted in a CB1R occupancy typically around ∼20-30%, thus much lower than what would be expected for classical G-protein coupled receptor (GPCR) antagonists in other therapeutic contexts. These findings are also discussed in relation to emerging literature on the potential usefulness of 'neutral' vs. 'classical' CB1R (inverse agonist) antagonists. The study additionally highlighted the usefulness of the radioligand [(11)C]SD5024 as a specific tracer for CB1R in the primate brain, though an arterial input function would ideally be required in future studies to further assure accurate quantitative analysis of specific binding.
Collapse
Affiliation(s)
- Stephan Hjorth
- Biosciences, CVMD Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Cecilia Karlsson
- CVMD Translational Medicine Unit, Early Clinical Development, Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden.
| | - Aurelija Jucaite
- AstraZeneca Translational Science Centre and Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Varnäs
- Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Wählby Hamrén
- Quantitative Clinical Pharmacology, Early Clinical Development, Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Peter Johnström
- AstraZeneca Translational Science Centre and Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Balázs Gulyás
- Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sean R Donohue
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christer Halldin
- Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lars Farde
- AstraZeneca Translational Science Centre and Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
|
41
|
Joshi EM, Need A, Schaus J, Chen Z, Benesh D, Mitch C, Morton S, Raub TJ, Phebus L, Barth V. Efficiency gains in tracer identification for nuclear imaging: can in vivo LC-MS/MS evaluation of small molecules screen for successful PET tracers? ACS Chem Neurosci 2014; 5:1154-63. [PMID: 25247893 DOI: 10.1021/cn500073j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Positron emission tomography (PET) imaging has become a useful noninvasive technique to explore molecular biology within living systems; however, the utility of this method is limited by the availability of suitable radiotracers to probe specific targets and disease biology. Methods to identify potential areas of improvement in the ability to predict small molecule performance as tracers prior to radiolabeling would speed the discovery of novel tracers. In this retrospective analysis, we characterized the brain penetration or peak SUV (standardized uptake value), binding potential (BP), and brain exposure kinetics across a series of known, nonradiolabeled PET ligands using in vivo LC-MS/MS (liquid chromatography coupled to mass spectrometry) and correlated these parameters with the reported PET ligand performance in nonhuman primates and humans available in the literature. The PET tracers studied included those reported to label G protein-coupled receptors (GPCRs), intracellular enzymes, and transporters. Additionally, data for each tracer was obtained from a mouse brain uptake assay (MBUA), previously published, where blood-brain barrier (BBB) penetration and clearance parameters were assessed and compared against similar data collected on a broad compound set of central nervous system (CNS) therapeutic compounds. The BP and SUV identified via nonradiolabeled LC-MS/MS, while different from the published values observed in the literature PET tracer data, allowed for an identification of initial criteria values we sought to facilitate increased potential for success from our early discovery screening paradigm. Our analysis showed that successful, as well as novel, clinical PET tracers exhibited BP of greater than 1.5 and peak SUVs greater than approximately 150% at 5 min post dose in rodents. The brain kinetics appeared similar between both techniques despite differences in tracer dose, suggesting linearity across these dose ranges. The assessment of tracers in a CNS exposure model, the mouse brain uptake assessment (MBUA), showed that those compound with initial brain-to-plasma ratios >2 and unbound fraction in brain homogenate >0.01 were more likely to be clinically successful PET ligands. Taken together, early incorporation of a LC/MS/MS cold tracer discovery assay and a parallel MBUA can be an useful screening paradigm to prioritize and rank order potential novel PET radioligands during early tracer discovery efforts. Compounds considered for continued in vivo PET assessments can be identified quickly by leveraging in vitro affinity and selectivity measures, coupled with data from a MBUA, primarily the 5 min brain-to-plasma ratio and unbound fraction data. Coupled utilization of these data creates a strategy to efficiently screen for the identification of appropriate chemical space to invest in for radiotracer discovery.
Collapse
Affiliation(s)
- Elizabeth M. Joshi
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Anne Need
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - John Schaus
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Zhaogen Chen
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Dana Benesh
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Charles Mitch
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Stuart Morton
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Thomas J. Raub
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Lee Phebus
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Vanessa Barth
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| |
Collapse
|
42
|
D’Addario C, Micioni Di Bonaventura M, Pucci M, Romano A, Gaetani S, Ciccocioppo R, Cifani C, Maccarrone M. Endocannabinoid signaling and food addiction. Neurosci Biobehav Rev 2014; 47:203-24. [DOI: 10.1016/j.neubiorev.2014.08.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 07/28/2014] [Accepted: 08/18/2014] [Indexed: 10/24/2022]
|
43
|
Thomae D, Morley TJ, Hamill T, Carroll VM, Papin C, Twardy NM, Lee HS, Hargreaves R, Baldwin RM, Tamagnan G, Alagille D. Automated one-step radiosynthesis of the CB1 receptor imaging agent [(18) F]MK-9470. J Labelled Comp Radiopharm 2014; 57:611-4. [PMID: 25156811 DOI: 10.1002/jlcr.3219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/24/2014] [Accepted: 06/26/2014] [Indexed: 12/14/2022]
Abstract
The fluorine-18-labeled positron emission tomography (PET) radiotracer [(18) F]MK-9470 is a selective, high affinity inverse agonist that has been used to image the cannabinoid receptor type 1 in human brain in healthy and disease states. This report describes a simplified, one-step [(18) F]radiofluorination approach using a GE TRACERlab FXFN module for the routine production of this tracer. The one-step synthesis, by [(18) F]fluoride displacement of a primary tosylate precursor, gives a six-fold increase in yield over the previous two-step method employing O-alkylation of a phenol precursor with 1,2-[(18) F]fluorobromoethane. The average radiochemical yield of [(18) F]MK-9470 using the one-step method was 30.3 ± 11.7% (n = 12), with specific activity in excess of 6 Ci/µmol and radiochemical purity of 97.2 ± 1.5% (n = 12), in less than 60 min. This simplified, high yielding, automated process was validated for routine GMP production of [(18) F]MK-9470 for clinical studies.
Collapse
Affiliation(s)
- David Thomae
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Huntington’s disease (HD) is a progressive and fatal neurodegenerative disorder caused by an expanded trinucleotide CAG sequence in huntingtin gene (HTT) on chromosome 4. HD manifests with chorea, cognitive and psychiatric symptoms. Although advances in genetics allow identification of individuals carrying the HD gene, much is still unknown about the mechanisms underlying the development of overt clinical symptoms and the transitional period between premanifestation and manifestation of the disease. HD has no cure and patients rely only in symptomatic treatment. There is an urgent need to identify biomarkers that are able to monitor disease progression and assess the development and efficacy of novel disease modifying drugs. Over the past years, neuroimaging techniques such as magnetic resonance imaging (MRI) and positron emission tomography (PET) have provided important advances in our understanding of HD. MRI provides information about structural and functional organization of the brain, while PET can detect molecular changes in the brain. MRI and PET are able to detect changes in the brains of HD gene carriers years ahead of the manifestation of the disease and have also proved to be powerful in assessing disease progression. However, no single technique has been validated as an optimal biomarker. An integrative multimodal imaging approach, which combines different MRI and PET techniques, could be recommended for monitoring potential neuroprotective and preventive therapies in HD. In this article we review the current neuroimaging literature in HD.
Collapse
|
45
|
Niccolini F, Loane C, Politis M. Dyskinesias in Parkinson's disease: views from positron emission tomography studies. Eur J Neurol 2014; 21:694-9, e39-43. [PMID: 24471508 DOI: 10.1111/ene.12362] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/17/2013] [Indexed: 11/27/2022]
Abstract
Levodopa-induced dyskinesias (LIDs) and graft-induced dyskinesias (GIDs) are serious and common complications of Parkinson's disease (PD) management following chronic treatment with levodopa or intrastriatal transplantation with dopamine-rich foetal ventral mesencephalic tissue, respectively. Positron emission tomography (PET) molecular imaging provides a powerful in vivo tool that has been employed over the past 20 years for the elucidation of mechanisms underlying the development of LIDs and GIDs in PD patients. PET used together with radioligands tagging molecular targets has allowed the functional investigation of several systems in the brain including the dopaminergic, serotonergic, glutamatergic, opioid, endocannabinoid, noradrenergic and cholinergic systems. In this article the role of PET imaging in unveiling pathophysiological mechanisms underlying the development of LIDs and GIDs in PD patients is reviewed.
Collapse
Affiliation(s)
- F Niccolini
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK; Neurodegeneration Imaging Group, Department of Clinical Neuroscience, King's College London, London, UK
| | | | | |
Collapse
|
46
|
Riaño Barros DA, McGinnity CJ, Rosso L, Heckemann RA, Howes OD, Brooks DJ, Duncan JS, Turkheimer FE, Koepp MJ, Hammers A. Test-retest reproducibility of cannabinoid-receptor type 1 availability quantified with the PET ligand [¹¹C]MePPEP. Neuroimage 2014; 97:151-62. [PMID: 24736184 PMCID: PMC4283194 DOI: 10.1016/j.neuroimage.2014.04.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 11/30/2022] Open
Abstract
Background Endocannabinoids are involved in normal cognition, and dysfunction in cannabinoid-receptor-mediated neurotransmission has been suggested in a variety of neurological and psychiatric pathologies. The type 1 cannabinoid receptor (CB1) is widely expressed in the human central nervous system. The objective of this study was to quantify the test–retest reproducibility of measures of the PET ligand [11C]MePPEP in order to assess the stability of CB1-receptor quantification in humans in vivo. Methods Fifteen healthy subjects (eight females; median age 32 years, range 25 to 65 years) had a 90-minute PET scan on two occasions after injection of a median dose of [11C]MePPEP of 364 MBq. Metabolite-corrected arterial plasma input functions were obtained for all scans. Eight ROIs, reflecting different levels of receptor densities/concentrations, were defined automatically: hippocampus, anterior cingulate gyrus, inferior frontal gyrus, caudate nucleus, globus pallidus, nucleus accumbens, thalamus, and pons. We used seven quantification methods: reversible compartmental models with one and two tissue classes, two and four rate constants, and a variable blood volume term (2kbv; 4kbv); model-free (spectral) analyses with and without regularisation, including one with voxel-wise quantification; the simplified reference tissue model (SRTM) with pons as a pseudo-reference region; and modified standard uptake values (mSUVs) calculated for the period of ~ 30–60 min after injection. Percentage test–retest change and between-subject variability were both assessed, and test–retest reliability was quantified by the intraclass correlation coefficient (ICC). The ratio of binding estimates pallidum:pons served as an indicator of a method's ability to reflect binding heterogeneity. Results Neither the SRTM nor the 4kbv model produced reliable measures, with ICCs around zero. Very good (> 0.75) or excellent (> 0.80) ICCs were obtained with the other methods. The most reliable were spectral analysis parametric maps (average across regions ± standard deviation 0.83 ± 0.03), rank shaping regularised spectral analysis (0.82 ± 0.05), and the 2kbv model (0.82 ± 0.09), but mSUVs were also reliable for most regions (0.79 ± 0.13). Mean test–retest changes among the five well-performing methods ranged from 12 ± 10% for mSUVs to 16% for 2kbv. Intersubject variability was high, with mean between-subject coefficients of variation ranging from 32 ± 13% for mSUVs to 45% for 2kbv. The highest pallidum:pons ratios of binding estimates were achieved by mSUV (4.2), spectral analysis-derived parametric maps (3.6), and 2kbv (3.6). Conclusion Quantification of CB1 receptor availability using [11C]MePPEP shows good to excellent reproducibility with several kinetic models and model-free analyses, whether applied on a region-of-interest or voxelwise basis. Simple mSUV measures were also reliable for most regions, but do not allow fully quantitative interpretation. [11C]MePPEP PET is well placed as a tool to investigate CB1-receptor mediated neurotransmission in health and disease. [11C]MePPEP is a PET tracer for cannabinoid receptors (CB1R). Extensive evaluation of [11C]MePPEP data quantification strategies in large sample We highlight successful methods to quantify CB1R in regions of interest. Highly reliable parametric maps (ICC 0.83 ± 0.03) allow whole-brain surveys. Modified standard uptake values also reliable, without arterial input functions
Collapse
Affiliation(s)
- Daniela A Riaño Barros
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; MRC Clinical Sciences Centre Hammersmith Hospital, London, UK
| | - Colm J McGinnity
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; MRC Clinical Sciences Centre Hammersmith Hospital, London, UK
| | - Lula Rosso
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK
| | - Rolf A Heckemann
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Neurodis Foundation, CERMEP, Imagerie du Vivant, Lyon. France
| | - Oliver D Howes
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; MRC Clinical Sciences Centre Hammersmith Hospital, London, UK
| | - David J Brooks
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Institute of Clinical Medicine, Aarhus University, Denmark
| | - John S Duncan
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, UK; Epilepsy Society, Chalfont St Peter, UK
| | | | - Matthias J Koepp
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, UK; Epilepsy Society, Chalfont St Peter, UK
| | - Alexander Hammers
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; MRC Clinical Sciences Centre Hammersmith Hospital, London, UK; Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, UK; Epilepsy Society, Chalfont St Peter, UK; Neurodis Foundation, CERMEP, Imagerie du Vivant, Lyon. France.
| |
Collapse
|
47
|
Lohith TG, Zoghbi SS, Morse CL, Araneta MDF, Barth VN, Goebl NA, Tauscher JT, Pike VW, Innis RB, Fujita M. Retest imaging of [11C]NOP-1A binding to nociceptin/orphanin FQ peptide (NOP) receptors in the brain of healthy humans. Neuroimage 2014; 87:89-95. [PMID: 24225488 PMCID: PMC3928240 DOI: 10.1016/j.neuroimage.2013.10.068] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 09/26/2013] [Accepted: 10/31/2013] [Indexed: 11/27/2022] Open
Abstract
[(11)C]NOP-1A is a novel high-affinity PET ligand for imaging nociceptin/orphanin FQ peptide (NOP) receptors. Here, we report reproducibility and reliability measures of binding parameter estimates for [(11)C]NOP-1A binding in the brain of healthy humans. After intravenous injection of [(11)C]NOP-1A, PET scans were conducted twice on eleven healthy volunteers on the same (10/11 subjects) or different (1/11 subjects) days. Subjects underwent serial sampling of radial arterial blood to measure parent radioligand concentrations. Distribution volume (VT; a measure of receptor density) was determined by compartmental (one- and two-tissue) modeling in large regions and by simpler regression methods (graphical Logan and bilinear MA1) in both large regions and voxel data. Retest variability and intraclass correlation coefficient (ICC) of VT were determined as measures of reproducibility and reliability respectively. Regional [(11)C]NOP-1A uptake in the brain was high, with a peak radioactivity concentration of 4-7 SUV (standardized uptake value) and a rank order of putamen>cingulate cortex>cerebellum. Brain time-activity curves fitted well in 10 of 11 subjects by unconstrained two-tissue compartmental model. The retest variability of VT was moderately good across brain regions except cerebellum, and was similar across different modeling methods, averaging 12% for large regions and 14% for voxel-based methods. The retest reliability of VT was also moderately good in most brain regions, except thalamus and cerebellum, and was similar across different modeling methods averaging 0.46 for large regions and 0.48 for voxels having gray matter probability >20%. The lowest retest variability and highest retest reliability of VT were achieved by compartmental modeling for large regions, and by the parametric Logan method for voxel-based methods. Moderately good reproducibility and reliability measures of VT for [(11)C]NOP-1A make it a useful PET ligand for comparing NOP receptor binding between different subject groups or under different conditions in the same subject.
Collapse
Affiliation(s)
- Talakad G Lohith
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Maria D Ferraris Araneta
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Masahiro Fujita
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
Hillard CJ, Liu QS. Endocannabinoid signaling in the etiology and treatment of major depressive illness. Curr Pharm Des 2014; 20:3795-811. [PMID: 24180398 PMCID: PMC4002665 DOI: 10.2174/13816128113196660735] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/23/2013] [Indexed: 12/28/2022]
Abstract
The purpose of this review is to examine human and preclinical data that are relevant to the following hypotheses. The first hypothesis is that deficient CB1R-mediated signaling results in symptoms that mimic those seen in depression. The second hypothesis is that activation of CB1R-mediated signaling results in behavioral, endocrine and other effects that are similar to those produced by currently used antidepressants. The third hypothesis is that conventional antidepressant therapies act through enhanced CB1R mediated signaling. Together the available data indicate that activators of CB1R signaling, particularly inhibitors of fatty acid amide hydrolase, should be considered for clinical trials for the treatment of depression.
Collapse
MESH Headings
- Animals
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
- Brain/drug effects
- Brain/enzymology
- Brain/metabolism
- Brain/pathology
- Cannabis
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Major/etiology
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/pathology
- Disease Models, Animal
- Endocannabinoids/genetics
- Endocannabinoids/metabolism
- Humans
- Magnetic Resonance Imaging
- Neurogenesis/drug effects
- Polymorphism, Genetic
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
| | - Qing-song Liu
- Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226.
| |
Collapse
|
49
|
Ceccarini J, De Hert M, Van Winkel R, Peuskens J, Bormans G, Kranaster L, Enning F, Koethe D, Leweke FM, Van Laere K. Increased ventral striatal CB1 receptor binding is related to negative symptoms in drug-free patients with schizophrenia. Neuroimage 2013; 79:304-12. [DOI: 10.1016/j.neuroimage.2013.04.052] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 03/26/2013] [Accepted: 04/15/2013] [Indexed: 11/16/2022] Open
|
50
|
Tsujikawa T, Zoghbi SS, Hong J, Donohue SR, Jenko KJ, Gladding RL, Halldin C, Pike VW, Innis RB, Fujita M. In vitro and in vivo evaluation of (11)C-SD5024, a novel PET radioligand for human brain imaging of cannabinoid CB1 receptors. Neuroimage 2013; 84:733-41. [PMID: 24076222 DOI: 10.1016/j.neuroimage.2013.09.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/04/2013] [Accepted: 09/18/2013] [Indexed: 10/26/2022] Open
Abstract
We recently developed a novel cannabinoid subtype-1 (CB1) receptor radioligand (11)C-SD5024 for brain imaging. This study aimed to evaluate (11)C-SD5024 both in vitro and in vivo and compare it with the other CB1 receptor ligands previously used in humans, i.e., (11)C-MePPEP, (11)C-OMAR, (18)F-MK-9470, and (18)F-FMPEP-d2. In vitro experiments were performed to measure dissociation constant (Ki) in the human brain and to measure the lipophilicity of the five CB1 receptor ligands listed above. In vivo specific binding in monkeys was measured by comparing total distribution volume (VT) at baseline and after full receptor blockade. The kinetics of (11)C-SD5024 in humans were evaluated in seven healthy subjects with compartmental modeling. SD5024 showed Ki=0.47nM, which was at an intermediate level among the five CB1 receptor ligands. Lipophilicity (LogD7.4) was 3.79, which is appropriate for brain imaging. Monkey scans showed high proportion of specific binding: ~80% of VT. In humans, (11)C-SD5024 showed peak brain uptake of 1.5-3 standardized uptake value, which was slightly higher than that of (11)C-OMAR and (18)F-MK-9470. One-compartment model showed good fitting, consistent with the vast majority of brain uptake being specific binding found in the monkey. Regional VT values were consistent with known distribution of CB1 receptors. VT calculated from 80 and 120min of scan data was strongly correlated (R(2)=0.97), indicating that 80min provided adequate information for quantitation and that the influence of radiometabolites was low. Intersubject variability for VT of (11)C-SD5024 was 22%, which was low among the five radioligands and indicated precise measurement. In conclusion, (11)C-SD5024 has appropriate affinity and lipophilicity, high specific binding, moderate brain uptake, and provides good precision to measure the binding. The results suggest that (11)C-SD5024 is slightly better than or equivalent to (11)C-OMAR and that both are suitable for clinical studies, especially those that involve two scans in one day.
Collapse
Affiliation(s)
- Tetsuya Tsujikawa
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|