1
|
Gärde M, Matheson GJ, Varnäs K, Svenningsson P, Hedman-Lagerlöf E, Lundberg J, Farde L, Tiger M. Altered Serotonin 1B Receptor Binding After Escitalopram for Depression Is Correlated With Treatment Effect. Int J Neuropsychopharmacol 2024; 27:pyae021. [PMID: 38695786 PMCID: PMC11119883 DOI: 10.1093/ijnp/pyae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is commonly treated with selective serotonin reuptake inhibitors (SSRIs). SSRIs inhibit the serotonin transporter (5-HTT), but the downstream antidepressant mechanism of action of these drugs is poorly understood. The serotonin 1B (5-HT1B) receptor is functionally linked to 5-HTT and 5-HT1B receptor binding and 5-HT1B receptor mRNA is reduced in the raphe nuclei after SSRI administration in primates and rodents, respectively. The effect of SSRI treatment on 5-HT1B receptor binding in patients with MDD has not been examined previously. This positron emission tomography (PET) study aimed to quantify brain 5-HT1B receptor binding changes in vivo after SSRI treatment for MDD in relation to treatment effect. METHODS Eight unmedicated patients with moderate to severe MDD underwent PET with the 5-HT1B receptor radioligand [11C]AZ10419369 before and after 3 to 4 weeks of treatment with the SSRI escitalopram 10 mg daily. Depression severity was assessed at time of PET and after 6 to 7 weeks of treatment with the Montgomery-Åsberg Depression Rating Scale. RESULTS We observed a significant reduction in [11C]AZ10419369 binding in a dorsal brainstem (DBS) region containing the median and dorsal raphe nuclei after escitalopram treatment (P = .036). Change in DBS [11C]AZ10419369 binding correlated with Montgomery-Åsberg Depression Rating Scale reduction after 3-4 (r = 0.78, P = .021) and 6-7 (r = 0.94, P < .001) weeks' treatment. CONCLUSIONS Our findings align with the previously reported reduction of 5-HT1B receptor binding in the raphe nuclei after SSRI administration and support future studies testing change in DBS 5-HT1B receptor binding as an SSRI treatment response marker.
Collapse
Affiliation(s)
- M Gärde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - G J Matheson
- Department of Psychiatry, Columbia University, New York, USA
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - K Varnäs
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - P Svenningsson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - E Hedman-Lagerlöf
- Division of Psychology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - J Lundberg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - L Farde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - M Tiger
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| |
Collapse
|
2
|
Fu H, Rong J, Chen Z, Zhou J, Collier T, Liang SH. Positron Emission Tomography (PET) Imaging Tracers for Serotonin Receptors. J Med Chem 2022; 65:10755-10808. [PMID: 35939391 DOI: 10.1021/acs.jmedchem.2c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) and 5-HT receptors (5-HTRs) have crucial roles in various neuropsychiatric disorders and neurodegenerative diseases, making them attractive diagnostic and therapeutic targets. Positron emission tomography (PET) is a noninvasive nuclear molecular imaging technique and is an essential tool in clinical diagnosis and drug discovery. In this context, numerous PET ligands have been developed for "visualizing" 5-HTRs in the brain and translated into human use to study disease mechanisms and/or support drug development. Herein, we present a comprehensive repertoire of 5-HTR PET ligands by focusing on their chemotypes and performance in PET imaging studies. Furthermore, this Perspective summarizes recent 5-HTR-focused drug discovery, including biased agonists and allosteric modulators, which would stimulate the development of more potent and subtype-selective 5-HTR PET ligands and thus further our understanding of 5-HTR biology.
Collapse
Affiliation(s)
- Hualong Fu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Zhen Chen
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Jingyin Zhou
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Thomas Collier
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
3
|
Rischka L, Vraka C, Pichler V, Rasul S, Nics L, Gryglewski G, Handschuh P, Murgaš M, Godbersen GM, Silberbauer LR, Unterholzner J, Wotawa C, Haider A, Ahmed H, Schibli R, Mindt T, Mitterhauser M, Wadsak W, Hahn A, Lanzenberger R, Hacker M, Ametamey SM. First-in-Humans Brain PET Imaging of the GluN2B-Containing N-methyl-d-aspartate Receptor with ( R)- 11C-Me-NB1. J Nucl Med 2022; 63:936-941. [PMID: 34620732 PMCID: PMC9157734 DOI: 10.2967/jnumed.121.262427] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/26/2021] [Indexed: 11/16/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) plays a crucial role in neurodegenerative diseases such as Alzheimer disease and in the treatment of major depression by fast-acting antidepressants such as ketamine. Given their broad implications, GluN2B-containing NMDARs have been of interest as diagnostic and therapeutic targets. Recently, (R)-11C-Me-NB1 was investigated preclinically and shown to be a promising radioligand for imaging GluN2B subunits. Here, we report on the performance characteristics of this radioligand in a first-in-humans PET study. Methods: Six healthy male subjects were scanned twice on a fully integrated PET/MR scanner with (R)-11C-Me-NB1 for 120 min. Brain uptake and tracer distribution over time were investigated by SUVs. Test-retest reliability was assessed with the absolute percentage difference and the coefficient of variation. Exploratory total volumes of distribution (VT) were computed using an arterial input function and the Logan plot as well as a constrained 2-tissue-compartment model with the ratio of rate constants between plasma and tissue compartments (K1/k2) coupled (2TCM). SUV was correlated with VT to investigate its potential as a surrogate marker of GluN2B expression. Results: High and heterogeneous radioligand uptake was observed across the entire gray matter with reversible kinetics within the scan time. SUV absolute percentage difference ranged from 6.9% to 8.5% and coefficient of variation from 4.9% to 6.0%, indicating a high test-retest reliability. A moderate correlation was found between SUV averaged from 70 to 90 min and VT using Logan plot (Spearman ρ = 0.44). Correlation between VT Logan and 2TCM was r = 0.76. Conclusion: The radioligand (R)-11C-Me-NB1 was highly effective in mapping GluN2B-enriched NMDARs in the human brain. With a heterogeneous uptake and a high test-retest reliability, this radioligand offers promise to deepen our understanding of the GluN2B-containing NMDAR in the pathophysiology and treatment of neuropsychiatric disease such as Alzheimer disease and major depression. Additionally, it could help in the selection of appropriate doses of GluN2B-targeting drugs.
Collapse
Affiliation(s)
- Lucas Rischka
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Chrysoula Vraka
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Verena Pichler
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Sazan Rasul
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Patricia Handschuh
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Matej Murgaš
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Godber M Godbersen
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Leo R Silberbauer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Jakob Unterholzner
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Christoph Wotawa
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Ahmed Haider
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland
| | - Hazem Ahmed
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland
| | - Roger Schibli
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland
| | - Thomas Mindt
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
- Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria; and
| | - Markus Mitterhauser
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Wolfgang Wadsak
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria;
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria;
| | - Simon M Ametamey
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland;
| |
Collapse
|
4
|
Mangeant R, Dubost E, Cailly T, Collot V. Radiotracers for the Central Serotoninergic System. Pharmaceuticals (Basel) 2022; 15:571. [PMID: 35631397 PMCID: PMC9143978 DOI: 10.3390/ph15050571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
This review lists the most important radiotracers described so far for imaging the central serotoninergic system. Single-photon emission computed tomography and positron emission tomography radiotracers are reviewed and critically discussed for each receptor.
Collapse
Affiliation(s)
- Reynald Mangeant
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| | - Emmanuelle Dubost
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| | - Thomas Cailly
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
- UNICAEN, IMOGERE, Normandie Univ., 14000 Caen, France
- CHU Côte de Nacre, Department of Nuclear Medicine, 14000 Caen, France
| | - Valérie Collot
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| |
Collapse
|
5
|
Veldman ER, Varrone A, Varnäs K, Svedberg MM, Cselényi Z, Tiger M, Gulyás B, Halldin C, Lundberg J. Serotonin 1B receptor density mapping of the human brainstem using positron emission tomography and autoradiography. J Cereb Blood Flow Metab 2022; 42:630-641. [PMID: 34644198 PMCID: PMC8943614 DOI: 10.1177/0271678x211049185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The serotonin 1B (5-HT1B) receptor has lately received considerable interest in relation to psychiatric and neurological diseases, partly due to findings based on quantification using Positron Emission Tomography (PET). Although the brainstem is an important structure in this regard, PET radioligand binding quantification in brainstem areas often shows poor reliability. This study aims to improve PET quantification of 5-HT1B receptor binding in the brainstem.Volumes of interest (VOIs) were selected based on a 3D [3H]AZ10419369 Autoradiography brainstem model, which visualized 5-HT1B receptor distribution in high resolution. Two previously developed VOI delineation methods were tested and compared to a conventional manual method. For a method based on template data, a [11C]AZ10419369 PET template was created by averaging parametric binding potential (BPND) images of 52 healthy subjects. VOIs were generated based on a predefined volume and BPND thresholding and subsequently applied to test-retest [11C]AZ10419369 parametric BPND images of 8 healthy subjects. For a method based on individual subject data, VOIs were generated directly on each individual parametric image.Both methods showed improved reliability compared to a conventional manual VOI. The VOIs created with [11C]AZ10419369 template data can be automatically applied to future PET studies measuring 5-HT1B receptor binding in the brainstem.
Collapse
Affiliation(s)
- Emma R Veldman
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Katarina Varnäs
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Marie M Svedberg
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.,Department of Health Promotion Science, Sophiahemmet University, Stockholm, Sweden
| | - Zsolt Cselényi
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.,PET Science Centre, Personalized Medicine and Biosamples, R&D, AstraZeneca, Stockholm, Sweden
| | - Mikael Tiger
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Balázs Gulyás
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Johan Lundberg
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
6
|
Svensson JE, Schain M, Knudsen GM, Ogden RT, Plavén-Sigray P. Early stopping in clinical PET studies: How to reduce expense and exposure. J Cereb Blood Flow Metab 2021; 41:2805-2819. [PMID: 34018825 PMCID: PMC8545054 DOI: 10.1177/0271678x211017796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/13/2021] [Accepted: 04/18/2021] [Indexed: 11/17/2022]
Abstract
Clinical positron emission tomography (PET) research is costly and entails exposing participants to radioactivity. Researchers should therefore aim to include just the number of subjects needed to fulfill the purpose of the study. In this tutorial we show how to apply sequential Bayes Factor testing in order to stop the recruitment of subjects in a clinical PET study as soon as enough data have been collected to make a conclusion. By using simulations, we demonstrate that it is possible to stop a study early, while keeping the number of erroneous conclusions low. We then apply sequential Bayes Factor testing to a real PET data set and show that it is possible to obtain support in favor of an effect while simultaneously reducing the sample size with 30%. Using this procedure allows researchers to reduce expense and radioactivity exposure for a range of effect sizes relevant for PET research.
Collapse
Affiliation(s)
- Jonas E Svensson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stockholm Health Care Services, Region Stockholm, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Schain
- Neurobiology Research Unit, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital, Copenhagen, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - R Todd Ogden
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA
| | - Pontus Plavén-Sigray
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stockholm Health Care Services, Region Stockholm, Karolinska University Hospital, Stockholm, Sweden
- Neurobiology Research Unit, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
7
|
Tjerkaski J, Cervenka S, Farde L, Matheson GJ. Kinfitr - an open-source tool for reproducible PET modelling: validation and evaluation of test-retest reliability. EJNMMI Res 2020; 10:77. [PMID: 32642865 PMCID: PMC7343683 DOI: 10.1186/s13550-020-00664-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/25/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In positron emission tomography (PET) imaging, binding is typically estimated by fitting pharmacokinetic models to the series of measurements of radioactivity in the target tissue following intravenous injection of a radioligand. However, there are multiple different models to choose from and numerous analytical decisions that must be made when modelling PET data. Therefore, it is important that analysis tools be adapted to the specific circumstances, and that analyses be documented in a transparent manner. Kinfitr, written in the open-source programming language R, is a tool developed for flexible and reproducible kinetic modelling of PET data, i.e. performing all steps using code which can be publicly shared in analysis notebooks. In this study, we compared outcomes obtained using kinfitr with those obtained using PMOD: a widely used commercial tool. RESULTS Using previously collected test-retest data obtained with four different radioligands, a total of six different kinetic models were fitted to time-activity curves derived from different brain regions. We observed good correspondence between the two kinetic modelling tools both for binding estimates and for microparameters. Likewise, no substantial differences were observed in the test-retest reliability estimates between the two tools. CONCLUSIONS In summary, we showed excellent agreement between the open-source R package kinfitr, and the widely used commercial application PMOD. We, therefore, conclude that kinfitr is a valid and reliable tool for kinetic modelling of PET data.
Collapse
Affiliation(s)
- Jonathan Tjerkaski
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden.
| | - Simon Cervenka
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Lars Farde
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Granville James Matheson
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| |
Collapse
|
8
|
Deen M, Hougaard A, Hansen HD, Schain M, Dyssegaard A, Knudsen GM, Ashina M. Association Between Sumatriptan Treatment During a Migraine Attack and Central 5-HT1B Receptor Binding. JAMA Neurol 2020; 76:834-840. [PMID: 31135819 DOI: 10.1001/jamaneurol.2019.0755] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Importance Triptans, the most efficient acute treatment for migraine attacks, are 5-HT1B/1D receptor agonists, but their precise mechanism of action is not completely understood. The extent to which triptans enter the central nervous system and bind to 5-HT1B receptors in the brain is unknown. Objectives To determine the occupancy of sumatriptan to central 5-HT1B receptors, and to investigate changes in brain serotonin levels during migraine attacks. Design, Setting, and Participants This study of 8 patients in Denmark used a within-participant design and was conducted from April 20, 2015, to December 5, 2016. Participants were otherwise healthy patients with untreated episodic migraine without aura, aged between 18 and 65 years, and recruited from the general community. Data analysis was performed from January 2017 to April 2018. Interventions All participants underwent positron emission tomographic scans after injection of [11C]AZ10419369, a specific 5-HT1B receptor radiotracer. All participants were scanned 3 times: (1) during an experimentally induced migraine attack, (2) after a subcutaneous injection of 6-mg subcutaneous sumatriptan, and (3) on a migraine attack-free day. Scans 1 and 2 were conducted on the same study day. Each scan lasted for 90 minutes. Main Outcome and Measure The primary outcome was the nondisplaceable binding potential of [11C]AZ10419369 across 7 brain regions involved in pain modulation. The binding potential reflects receptor density, and changes in binding potential reflects displacement of the radiotracer. The occupancy of sumatriptan was estimated from the 2 scans before and after sumatriptan administration. Results Eight patients with migraine were included in the study; of these participants, 7 (87%) were women. The mean (SD) age of participants on study day 1 was 29.5 (9.2) years and on study day 2 was 30.0 (8.9) years. Sumatriptan was associated with statistically significantly reduced 5-HT1B receptor binding across pain-modulating regions (mean [SD] binding potential, 1.20 [0.20] vs 1.02 [0.22]; P = .001), corresponding to a mean (SD) drug occupancy rate of 16.0% (5.3%). Furthermore, during migraine attacks, as compared with outside of attacks, 5-HT1B receptor binding was statistically significantly associated with reduced in pain-modulating regions (mean [SD] binding potential, 1.36 [0.22] vs 1.20 [0.20]; P = .02). Conclusions and Relevance Treatment with sumatriptan during migraine attacks appeared to be associated with a decrease in 5-HT1B receptor binding, a finding that is most likely associated with the binding of sumatriptan to central 5-HT1B receptors, but the contribution of ongoing cerebral serotonin release to the lower binding cannot be excluded; the migraine attack-associated decrease in binding could indicate that migraine attacks are associated with increases in endogenous serotonin.
Collapse
Affiliation(s)
- Marie Deen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup, Denmark.,Neurobiology Research Unit and NeuroPharm, Department of Neurology, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Hougaard
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup, Denmark
| | - Hanne Demant Hansen
- Neurobiology Research Unit and NeuroPharm, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Martin Schain
- Neurobiology Research Unit and NeuroPharm, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Agnete Dyssegaard
- Neurobiology Research Unit and NeuroPharm, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Gitte Moos Knudsen
- Neurobiology Research Unit and NeuroPharm, Department of Neurology, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Cselényi Z, Jucaite A, Kristensson C, Stenkrona P, Ewing P, Varrone A, Johnström P, Schou M, Vazquez-Romero A, Moein MM, Bolin M, Siikanen J, Grybäck P, Larsson B, Halldin C, Grime K, Eriksson UG, Farde L. Quantification and reliability of [ 11C]VC - 002 binding to muscarinic acetylcholine receptors in the human lung - a test-retest PET study in control subjects. EJNMMI Res 2020; 10:59. [PMID: 32495011 PMCID: PMC7270393 DOI: 10.1186/s13550-020-00634-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/22/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The radioligand [11C]VC-002 was introduced in a small initial study long ago for imaging of muscarinic acetylcholine receptors (mAChRs) in human lungs using positron emission tomography (PET). The objectives of the present study in control subjects were to advance the methodology for quantification of [11C]VC-002 binding in lung and to examine the reliability using a test-retest paradigm. This work constituted a self-standing preparatory step in a larger clinical trial aiming at estimating mAChR occupancy in the human lungs following inhalation of mAChR antagonists. METHODS PET measurements using [11C]VC-002 and the GE Discovery 710 PET/CT system were performed in seven control subjects at two separate occasions, 2-19 days apart. One subject discontinued the study after the first measurement. Radioligand binding to mAChRs in lung was quantified using an image-derived arterial input function. The total distribution volume (VT) values were obtained on a regional and voxel-by-voxel basis. Kinetic one-tissue and two-tissue compartment models (1TCM, 2TCM), analysis based on linearization of the compartment models (multilinear Logan) and image analysis by data-driven estimation of parametric images based on compartmental theory (DEPICT) were applied. The test-retest repeatability of VT estimates was evaluated by absolute variability (VAR) and intraclass correlation coefficients (ICCs). RESULTS The 1TCM was the statistically preferred model for description of [11C]VC-002 binding in the lungs. Low VAR (< 10%) across analysis methods indicated good reliability of the PET measurements. The VT estimates were stable after 60 min. CONCLUSIONS The kinetic behaviour and good repeatability of [11C]VC-002 as well as the novel lung image analysis methodology support its application in applied studies on drug-induced mAChR receptor occupancy and the pathophysiology of pulmonary disorders. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03097380, registered: 31 March 2017.
Collapse
Affiliation(s)
- Zsolt Cselényi
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden.
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.
| | - Aurelija Jucaite
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | - Per Stenkrona
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Pär Ewing
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Peter Johnström
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Magnus Schou
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Ana Vazquez-Romero
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Mohammad Mahdi Moein
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Martin Bolin
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jonathan Siikanen
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Pär Grybäck
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Bengt Larsson
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Ken Grime
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | | | - Lars Farde
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
10
|
A randomized placebo-controlled PET study of ketamine´s effect on serotonin 1B receptor binding in patients with SSRI-resistant depression. Transl Psychiatry 2020; 10:159. [PMID: 32475989 PMCID: PMC7261801 DOI: 10.1038/s41398-020-0844-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 04/26/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
The glutamate N-methyl-D-aspartate receptor antagonist ketamine has a rapid antidepressant effect. Despite large research efforts, ketamine's mechanism of action in major depressive disorder (MDD) has still not been determined. In rodents, the antidepressant properties of ketamine were found to be dependent on both the α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) and the serotonin (5-HT)1B receptor. Low 5-HT1B receptor binding in limbic brain regions is a replicated finding in MDD. In non-human primates, AMPA-dependent increase in 5-HT1B receptor binding in the ventral striatum (VST) has been demonstrated after ketamine infusion. Thirty selective serotonin reuptake inhibitor-resistant MDD patients were recruited via advertisement and randomized to double-blind monotherapy with 0.5 mg/kg ketamine or placebo infusion. The patients were examined with the 5-HT1B receptor selective radioligand [11C]AZ10419369 and positron emission tomography (PET) before and 24-72 h after treatment. 5-HT1B receptor binding did not significantly alter in patients treated with ketamine compared with placebo. An increase in 5-HT1B receptor binding with 16.7 % (p = 0.036) was found in the hippocampus after one ketamine treatment. 5-HT1B receptor binding in VST at baseline correlated with MDD symptom ratings (r = -0.426, p = 0.019) and with reduction of depressive symptoms with ketamine (r = -0.644, p = 0.002). In conclusion, reduction of depressive symptoms in MDD patients after ketamine treatment is correlated inversely with baseline 5-HT1B receptor binding in VST. Further studies examining the role of 5-HT1B receptors in the antidepressant mechanism of action of ketamine should be conducted, homing in on the 5-HT1B receptor as an MDD treatment response marker.
Collapse
|
11
|
Matheson GJ. We need to talk about reliability: making better use of test-retest studies for study design and interpretation. PeerJ 2019; 7:e6918. [PMID: 31179173 PMCID: PMC6536112 DOI: 10.7717/peerj.6918] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 04/07/2019] [Indexed: 12/31/2022] Open
Abstract
Neuroimaging, in addition to many other fields of clinical research, is both time-consuming and expensive, and recruitable patients can be scarce. These constraints limit the possibility of large-sample experimental designs, and often lead to statistically underpowered studies. This problem is exacerbated by the use of outcome measures whose accuracy is sometimes insufficient to answer the scientific questions posed. Reliability is usually assessed in validation studies using healthy participants, however these results are often not easily applicable to clinical studies examining different populations. I present a new method and tools for using summary statistics from previously published test-retest studies to approximate the reliability of outcomes in new samples. In this way, the feasibility of a new study can be assessed during planning stages, and before collecting any new data. An R package called relfeas also accompanies this article for performing these calculations. In summary, these methods and tools will allow researchers to avoid performing costly studies which are, by virtue of their design, unlikely to yield informative conclusions.
Collapse
Affiliation(s)
- Granville J. Matheson
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
12
|
Stenkrona P, Matheson GJ, Cervenka S, Sigray PP, Halldin C, Farde L. [ 11C]SCH23390 binding to the D 1-dopamine receptor in the human brain-a comparison of manual and automated methods for image analysis. EJNMMI Res 2018; 8:74. [PMID: 30069645 PMCID: PMC6070454 DOI: 10.1186/s13550-018-0416-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 11/22/2022] Open
Abstract
Background The D1-dopamine receptor radioligand [11C]SCH23390 has been frequently used in PET studies. In drug-naïve patients with schizophrenia, the findings have been inconsistent, with decreases, increases, and no change in the frontal cortex D1-dopamine receptors. While these discrepancies are likely primarily due to a lack of statistical power in these studies, we speculated that an additional explanation may be the differences due to methods of image analysis between studies, affecting reliability as well as bias between groups. Methods Fifteen healthy subjects underwent two PET measurements with [11C]SCH23390 on the same day. The binding potential (BPND) was compared using a 95% confidence interval following manual and automated delineation of a region of interest (ROI) as well as with and without frame-by-frame realignment. Results Automated target region delineation produced lower BPND values, while automated delineation of the reference region yielded higher BPND values. However, no significant differences were observed for repeatability using automated and manual delineation methods. Frame-by-frame realignment generated higher BPND values and improved repeatability. Conclusions The results suggest that the choice of ROI delineation method is not an important factor for reliability, whereas the improved results following movement correction confirm its importance in PET image analysis. Realignment is therefore especially important for measurements in patient populations such as schizophrenia or Parkinson’s disease, where motion artifacts may be more prevalent. Electronic supplementary material The online version of this article (10.1186/s13550-018-0416-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Per Stenkrona
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska University Hospital, Karolinska Institutet, R5:02, S-171 76, Stockholm, Sweden. .,Stockholm County Council, Stockholm, Sweden.
| | - Granville J Matheson
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska University Hospital, Karolinska Institutet, R5:02, S-171 76, Stockholm, Sweden.,Stockholm County Council, Stockholm, Sweden
| | - Simon Cervenka
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska University Hospital, Karolinska Institutet, R5:02, S-171 76, Stockholm, Sweden.,Stockholm County Council, Stockholm, Sweden
| | - Pontus Plavén Sigray
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska University Hospital, Karolinska Institutet, R5:02, S-171 76, Stockholm, Sweden.,Stockholm County Council, Stockholm, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska University Hospital, Karolinska Institutet, R5:02, S-171 76, Stockholm, Sweden.,Stockholm County Council, Stockholm, Sweden
| | - Lars Farde
- PET Science Centre, Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
Yang KC, Takano A, Halldin C, Farde L, Finnema SJ. Serotonin concentration enhancers at clinically relevant doses reduce [ 11C]AZ10419369 binding to the 5-HT 1B receptors in the nonhuman primate brain. Transl Psychiatry 2018; 8:132. [PMID: 30013068 PMCID: PMC6048172 DOI: 10.1038/s41398-018-0178-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 02/14/2018] [Accepted: 04/03/2018] [Indexed: 12/25/2022] Open
Abstract
The serotonin (5-HT) system plays an important role in the pathophysiology and treatment of several major psychiatric disorders. Currently, no suitable positron emission tomography (PET) imaging paradigm is available to assess 5-HT release in the living human brain. [11C]AZ10419369 binds to 5-HT1B receptors and is one of the most 5-HT-sensitive radioligands available. This study applied 5-HT concentration enhancers which can be safely studied in humans, and examined their effect on [11C]AZ10419369 binding at clinically relevant doses, including amphetamine (1 mg/kg), 3,4-methylenedioxymethamphetamine (MDMA; 1 mg/kg) or 5-hydroxy-L-tryptophan (5-HTP; 5 mg/kg). Twenty-six PET measurements (14 for amphetamine, 6 for MDMA and 6 for 5-HTP) using a bolus and constant infusion protocol were performed in four cynomolgus monkeys before or after drug administration. Binding potential (BPND) values were determined with the equilibrium method (integral interval: 63-123 min) using cerebellum as the reference region. BPND values were significantly decreased in several examined brain regions after administration of amphetamine (range: 19-31%), MDMA (16-25%) or 5-HTP (13-31%). Reductions in [11C]AZ10419369 binding were greater in striatum than cortical regions after administration of 5-HTP, while no prominent regional differences were found for amphetamine and MDMA. In conclusion, [11C]AZ10419369 binding is sensitive to changes in 5-HT concentration induced by amphetamine, MDMA or 5-HTP. The robust changes in BPND, following pretreatment drugs administered at clinically relevant doses, indicate that the applied PET imaging paradigms hold promise to be successfully used in future human studies.
Collapse
Affiliation(s)
- Kai-Chun Yang
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Farde
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Personalized Health Care and Biomarkers, AstraZeneca PET Science Center at Karolinska Institutet, Stockholm, Sweden
| | - Sjoerd J Finnema
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| |
Collapse
|
14
|
Finnema SJ, Nabulsi NB, Eid T, Detyniecki K, Lin SF, Chen MK, Dhaher R, Matuskey D, Baum E, Holden D, Spencer DD, Mercier J, Hannestad J, Huang Y, Carson RE. Imaging synaptic density in the living human brain. Sci Transl Med 2017; 8:348ra96. [PMID: 27440727 DOI: 10.1126/scitranslmed.aaf6667] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/24/2016] [Indexed: 12/26/2022]
Abstract
Chemical synapses are the predominant neuron-to-neuron contact in the central nervous system. Presynaptic boutons of neurons contain hundreds of vesicles filled with neurotransmitters, the diffusible signaling chemicals. Changes in the number of synapses are associated with numerous brain disorders, including Alzheimer's disease and epilepsy. However, all current approaches for measuring synaptic density in humans require brain tissue from autopsy or surgical resection. We report the use of the synaptic vesicle glycoprotein 2A (SV2A) radioligand [(11)C]UCB-J combined with positron emission tomography (PET) to quantify synaptic density in the living human brain. Validation studies in a baboon confirmed that SV2A is an alternative synaptic density marker to synaptophysin. First-in-human PET studies demonstrated that [(11)C]UCB-J had excellent imaging properties. Finally, we confirmed that PET imaging of SV2A was sensitive to synaptic loss in patients with temporal lobe epilepsy. Thus, [(11)C]UCB-J PET imaging is a promising approach for in vivo quantification of synaptic density with several potential applications in diagnosis and therapeutic monitoring of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Sjoerd J Finnema
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA.
| | - Nabeel B Nabulsi
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Tore Eid
- Department of Laboratory Medicine, Yale University, New Haven, CT 06520, USA
| | - Kamil Detyniecki
- Department of Neurology, Yale University, New Haven, CT 06520, USA
| | - Shu-Fei Lin
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Ming-Kai Chen
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Roni Dhaher
- Department of Laboratory Medicine, Yale University, New Haven, CT 06520, USA
| | - David Matuskey
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Evan Baum
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Daniel Holden
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Dennis D Spencer
- Department of Neurosurgery, Yale University, New Haven, CT 06520, USA
| | | | | | - Yiyun Huang
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Richard E Carson
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA. Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
15
|
Arakawa R, Stenkrona P, Takano A, Nag S, Maior RS, Halldin C. Test-retest reproducibility of [ 11C]-L-deprenyl-D 2 binding to MAO-B in the human brain. EJNMMI Res 2017. [PMID: 28634836 PMCID: PMC5478550 DOI: 10.1186/s13550-017-0301-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background [11C]-l-deprenyl-D2 is a positron emission tomography (PET) radioligand for measurement of the monoamine oxidase B (MAO-B) activity in vivo brain. The estimation of the test-retest reproducibility is important for accurate interpretation of PET studies. Results We performed two [11C]-l-deprenyl-D2 scans for six healthy subjects and evaluated the test-retest variability of this radioligand. MAO-B binding was quantified by two tissue compartment model (2TCM) with three rate constants (K1, k2, k3) using metabolite-corrected plasma radioactivity. The λk3 defined as (K1/k2) × k3 was also calculated. The correlation between MAO-B binding and age, and the effect of partial volume effect correction (PVEc) for the reproducibility were also estimated. %difference of k3 was 2.6% (medial frontal cortex) to 10.3% (hippocampus), and that of λk3 was 5.0% (thalamus) to 9.2% (cerebellum). Mean %difference of all regions were 5.3 and 7.0% in k3 and λk3, respectively. All regions showed below 10% variabilities except the hippocampus in k3 (10.3%). Intraclass correlation coefficient (ICC) of k3 was 0.78 (hippocampus) to 0.98 (medial frontal cortex), and that of λk3 was 0.78 (hippocampus) to 0.95 (thalamus). Mean ICC were 0.94 and 0.89 in k3 and λk3, respectively. The highest positive correlation with age was observed in the hippocampus, as r = 0.75 in k3 and 0.76 in λk3. After PVEc, mean %difference were 5.6 and 7.2% in k3 and λk3, respectively. Mean ICC were 0.92 and 0.90 for k3 and λk3, respectively. These values were almost the same as those before PVEc. Conclusions The present results indicate that k3 and λk3 of [11C]-l-deprenyl-D2 are reliable parameters for test-retest reproducibility with healthy subjects both before and after PVEc. The studies with patients of larger sample size are required for further clinical applications.
Collapse
Affiliation(s)
- Ryosuke Arakawa
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.
| | - Per Stenkrona
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Sangram Nag
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Rafael S Maior
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.,Primate Center and Laboratory of Neurosciences and Behavior, Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
16
|
Deen M, Hansen HD, Hougaard A, da Cunha-Bang S, Nørgaard M, Svarer C, Keller SH, Thomsen C, Ashina M, Knudsen GM. Low 5-HT 1B receptor binding in the migraine brain: A PET study. Cephalalgia 2017; 38:519-527. [PMID: 28730894 DOI: 10.1177/0333102417698708] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background The pathophysiology of migraine may involve dysfunction of serotonergic signaling. In particular, the 5-HT1B receptor is considered a key player due to the efficacy of 5-HT1B receptor agonists for treatment of migraine attacks. Aim To examine the cerebral 5-HT1B receptor binding in interictal migraine patients without aura compared to controls. Methods Eighteen migraine patients, who had been migraine free for >48 hours, and 16 controls were scanned after injection of the 5-HT1B receptor specific radioligand [11C]AZ10419369 for quantification of cerebral 5-HT1B receptor binding. Patients who reported migraine <48 hours after the PET examination were excluded from the final analysis. We defined seven brain regions involved in pain modulation as regions of interest and applied a latent variable model (LVM) to assess the group effect on binding across these regions. Results Our data support a model wherein group status predicts the latent variable ( p = 0.038), with migraine patients having lower 5-HT1B receptor binding across regions compared to controls. Further, in a whole-brain voxel-based analysis, time since last migraine attack correlated positively with 5-HT1B receptor binding in the dorsal raphe and in the midbrain. Conclusion We report here for the first time that migraine patients have low 5-HT1B receptor binding in pain modulating regions, reflecting decreased receptor density. This is either a primary constitutive trait of the migraine brain or secondary to repeated exposure to migraine attacks. We also provide indirect support for the dorsal raphe 5-HT1B receptors being temporarily downregulated during the migraine attack, presumably in response to higher cerebral serotonin levels in the ictal phase.
Collapse
Affiliation(s)
- Marie Deen
- 1 Danish Headache Center and Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark.,2 Neurobiology Research Unit and Center for Experimental Medicine Neuropharmacology, Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark.,3 Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hanne D Hansen
- 2 Neurobiology Research Unit and Center for Experimental Medicine Neuropharmacology, Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark
| | - Anders Hougaard
- 1 Danish Headache Center and Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark
| | - Sofi da Cunha-Bang
- 2 Neurobiology Research Unit and Center for Experimental Medicine Neuropharmacology, Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark.,3 Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Nørgaard
- 2 Neurobiology Research Unit and Center for Experimental Medicine Neuropharmacology, Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark
| | - Claus Svarer
- 2 Neurobiology Research Unit and Center for Experimental Medicine Neuropharmacology, Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark
| | - Sune H Keller
- 4 Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Carsten Thomsen
- 5 Department of Radiology, Rigshospitalet, Copenhagen, Denmark
| | - Messoud Ashina
- 1 Danish Headache Center and Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark.,3 Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gitte M Knudsen
- 2 Neurobiology Research Unit and Center for Experimental Medicine Neuropharmacology, Department of Neurology, The Neuroscience Centre, Rigshospitalet, Denmark.,3 Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Tiger M, Farde L, Rück C, Varrone A, Forsberg A, Lindefors N, Halldin C, Lundberg J. Low serotonin1B receptor binding potential in the anterior cingulate cortex in drug-free patients with recurrent major depressive disorder. Psychiatry Res Neuroimaging 2016; 253:36-42. [PMID: 27269199 DOI: 10.1016/j.pscychresns.2016.04.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 12/17/2022]
Abstract
The pathophysiology of major depressive disorder (MDD) is not fully understood and the diagnosis is largely based on history and clinical examination. So far, several lines of preclinical data and a single imaging study implicate a role for the serotonin1B (5-HT1B) receptor subtype. We sought to study 5-HT1B receptor binding in brain regions of reported relevance in patients with MDD. Subjects were examined at the Karolinska Institutet PET centre using positron emission tomography (PET) and the 5-HT1B receptor selective radioligand [(11)C]AZ10419369. Ten drug-free patients with recurrent MDD and ten control subjects matched for age and sex were examined. The main outcome measure was [(11)C]AZ10419369 binding in brain regions of reported relevance in the pathophysiology of MDD. The [(11)C]AZ10419369 binding potential was significantly lower in the MDD group compared with the healthy control group in the anterior cingulate cortex (20% between-group difference), the subgenual prefrontal cortex (17% between-group difference), and in the hippocampus (32% between-group difference). The low anterior cingulate [(11)C]AZ10419369 binding potential in patients with recurrent MDD positions 5-HT1B receptor binding in this region as a putative biomarker for MDD and corroborate a role of the anterior cingulate cortex and associated areas in the pathophysiology of recurrent MDD.
Collapse
Affiliation(s)
- Mikael Tiger
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden.
| | - Lars Farde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden; AstraZeneca, Translational Science Center, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Christian Rück
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Andrea Varrone
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Anton Forsberg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Nils Lindefors
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Christer Halldin
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Johan Lundberg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| |
Collapse
|
18
|
Pittenger C, Adams TG, Gallezot JD, Crowley MJ, Nabulsi N, Ropchan J, Gao H, Kichuk SA, Simpson R, Billingslea E, Hannestad J, Bloch M, Mayes L, Bhagwagar Z, Carson RE. OCD is associated with an altered association between sensorimotor gating and cortical and subcortical 5-HT1b receptor binding. J Affect Disord 2016; 196:87-96. [PMID: 26919057 PMCID: PMC4808438 DOI: 10.1016/j.jad.2016.02.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/02/2015] [Accepted: 02/07/2016] [Indexed: 01/04/2023]
Abstract
Obsessive-compulsive disorder (OCD) is characterized by impaired sensorimotor gating, as measured using prepulse inhibition (PPI). This effect may be related to abnormalities in the serotonin (5-HT) system. 5-HT1B agonists can impair PPI, produce OCD-like behaviors in animals, and exacerbate OCD symptoms in humans. We measured 5-HT1B receptor availability using (11)C-P943 positron emission tomography (PET) in unmedicated, non-depressed OCD patients (n=12) and matched healthy controls (HC; n=12). Usable PPI data were obtained from 20 of these subjects (10 from each group). There were no significant main effects of OCD diagnosis on 5-HT1B receptor availability ((11)C-P943 BPND); however, the relationship between PPI and (11)C-P943 BPND differed dramatically and significantly between groups. 5-HT1B receptor availability in the basal ganglia and thalamus correlated positively with PPI in controls; these correlations were lost or even reversed in the OCD group. In cortical regions there were no significant correlations with PPI in controls, but widespread positive correlations in OCD patients. Positive correlations between 5-HT1B receptor availability and PPI were consistent across diagnostic groups only in two structures, the orbitofrontal cortex and the amygdala. Differential associations of 5-HT1B receptor availability with PPI in patients suggest functionally important alterations in the serotonergic regulation of cortical/subcortical balance in OCD.
Collapse
Affiliation(s)
- Christopher Pittenger
- Department of Psychiatry, Yale University School of Medicine, USA; Department of Psychology, Yale University School of Medicine, USA; Child Study Center, Yale University School of Medicine, USA; Interdepartmental Neuroscience Program, Yale University School of Medicine, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
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: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Kumar JSD, Mann JJ. PET tracers for serotonin receptors and their applications. Cent Nerv Syst Agents Med Chem 2015; 14:96-112. [PMID: 25360773 DOI: 10.2174/1871524914666141030124316] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/26/2014] [Accepted: 10/28/2014] [Indexed: 11/22/2022]
Abstract
Serotonin receptors (5-HTRs) are implicated in the pathophysiology of a variety of neuropsychiatric and neurodegenerative disorders and are also targets for drug therapy. In the CNS, most of these receptors are expressed in high abundance in specific brain regions reflecting their role in brain functions. Quantifying binding to 5-HTRs in vivo may permit assessment of physiologic and pathologic conditions, and monitoring disease progression, evaluating treatment response, and for investigating new treatment modalities. Positron emission tomography (PET) molecular imaging has the sensitivity to quantify binding of 5-HTRs in CNS disorders and to measure drug occupancy as part of a process of new drug development. Although research on PET imaging of 5-HTRs have been performed more than two decades, the successful radiotracers so far developed for human studies are limited to 5-HT₁AR, 5-HT₁BR, 5-HT₂AR, 5-HT₄R and 5-HT₆R. Herein we review the development and application of radioligands for PET imaging of 5-HTRs in living brain.
Collapse
Affiliation(s)
| | - J John Mann
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric institute, 1051 Riverside Drive, Box: 42, New York, NY, 10032, USA.
| |
Collapse
|
21
|
Finnema SJ, Scheinin M, Shahid M, Lehto J, Borroni E, Bang-Andersen B, Sallinen J, Wong E, Farde L, Halldin C, Grimwood S. Application of cross-species PET imaging to assess neurotransmitter release in brain. Psychopharmacology (Berl) 2015; 232:4129-57. [PMID: 25921033 PMCID: PMC4600473 DOI: 10.1007/s00213-015-3938-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/09/2015] [Indexed: 01/03/2023]
Abstract
RATIONALE This review attempts to summarize the current status in relation to the use of positron emission tomography (PET) imaging in the assessment of synaptic concentrations of endogenous mediators in the living brain. OBJECTIVES Although PET radioligands are now available for more than 40 CNS targets, at the initiation of the Innovative Medicines Initiative (IMI) "Novel Methods leading to New Medications in Depression and Schizophrenia" (NEWMEDS) in 2009, PET radioligands sensitive to an endogenous neurotransmitter were only validated for dopamine. NEWMEDS work-package 5, "Cross-species and neurochemical imaging (PET) methods for drug discovery", commenced with a focus on developing methods enabling assessment of changes in extracellular concentrations of serotonin and noradrenaline in the brain. RESULTS Sharing the workload across institutions, we utilized in vitro techniques with cells and tissues, in vivo receptor binding and microdialysis techniques in rodents, and in vivo PET imaging in non-human primates and humans. Here, we discuss these efforts and review other recently published reports on the use of radioligands to assess changes in endogenous levels of dopamine, serotonin, noradrenaline, γ-aminobutyric acid, glutamate, acetylcholine, and opioid peptides. The emphasis is on assessment of the availability of appropriate translational tools (PET radioligands, pharmacological challenge agents) and on studies in non-human primates and human subjects, as well as current challenges and future directions. CONCLUSIONS PET imaging directed at investigating changes in endogenous neurochemicals, including the work done in NEWMEDS, have highlighted an opportunity to further extend the capability and application of this technology in drug development.
Collapse
Affiliation(s)
- Sjoerd J. Finnema
- />Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Mika Scheinin
- />Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland , />Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Mohammed Shahid
- />Research and Development, Orion Corporation, Orion Pharma, Turku, Finland
| | - Jussi Lehto
- />Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland
| | - Edilio Borroni
- />Neuroscience Department, Hoffman-La Roche, Basel, Switzerland
| | | | - Jukka Sallinen
- />Research and Development, Orion Corporation, Orion Pharma, Turku, Finland
| | - Erik Wong
- />Neuroscience Innovative Medicine Unit, AstraZeneca, Wilmington, DE USA
| | - Lars Farde
- />Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden , />Translational Science Center at Karolinska Institutet, AstraZeneca, Stockholm, Sweden
| | - Christer Halldin
- />Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Sarah Grimwood
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, USA. .,, 610 Main Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
22
|
Saricicek A, Chen J, Planeta B, Ruf B, Subramanyam K, Maloney K, Matuskey D, Labaree D, Deserno L, Neumeister A, Krystal JH, Gallezot JD, Huang Y, Carson RE, Bhagwagar Z. Test-retest reliability of the novel 5-HT1B receptor PET radioligand [11C]P943. Eur J Nucl Med Mol Imaging 2014; 42:468-77. [PMID: 25427881 DOI: 10.1007/s00259-014-2958-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 11/03/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE [(11)C]P943 is a novel, highly selective 5-HT1B PET radioligand. The aim of this study was to determine the test-retest reliability of [(11)C]P943 using two different modeling methods and to perform a power analysis with each quantification technique. METHODS Seven healthy volunteers underwent two PET scans on the same day. Regions of interest (ROIs) were the amygdala, hippocampus, pallidum, putamen, insula, frontal, anterior cingulate, parietal, temporal and occipital cortices, and cerebellum. Two multilinear radioligand quantification techniques were used to estimate binding potential: MA1, using arterial input function data, and the second version of the multilinear reference tissue model analysis (MRTM2), using the cerebellum as the reference region. Between-scan percent variability and intraclass correlation coefficients (ICC) were used to assess test-retest reliability. We also performed power analyses to determine the method that would allow the least number of subjects using within-subject or between-subject study designs. A voxel-wise ICC analysis for MRTM2 BPND was performed for the whole brain and all the ROIs studied. RESULTS Mean percent variability between two scans across regions ranged between 0.4 % and 12.4 % for MA1 BPND, 0.5 % and 11.5 % for MA1 BPP, 16.7 % and 28.3 % for MA1 BPF, and between 0.2 % and 5.4 % for MRTM2 BPND. The power analyses showed a greater number of subjects were required using MA1 BPF compared with other outcome measures for both within-subject and between-subject study designs. ICC values were the highest using MRTM2 BPND and the lowest with MA1 BPF in ten ROIs. Small regions and regions with low binding had lower ICC values than large regions and regions with high binding. CONCLUSION Reliable measures of 5-HT1B receptor binding can be obtained using the novel PET radioligand [(11)C]P943. Quantification of 5-HT1B receptor binding with MRTM2 BPND and with MA1 BPP provided the least variability and optimal power for within-subject and between-subject designs.
Collapse
|
23
|
Nord M, Cselenyi Z, Forsberg A, Rosenqvist G, Tiger M, Lundberg J, Varrone A, Farde L. Distinct regional age effects on [11C]AZ10419369 binding to 5-HT1B receptors in the human brain. Neuroimage 2014; 103:303-308. [PMID: 25255943 DOI: 10.1016/j.neuroimage.2014.09.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/21/2014] [Accepted: 09/15/2014] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Age-related changes in the serotonin system have been described, and proposed to be associated with behavioral changes observed particularly in the elderly population. The 5-HT1B receptor is thought to have a regulatory role in a number of physiological functions, and has been implicated in several age-related diseases. The purpose of the present study was to examine if the availability of 5-HT1B receptors is decreasing with age in healthy subjects. METHODS Data from five previous studies were reanalyzed and pooled, generating data from fifty-one healthy subjects, age 20 to 70, that had been examined with positron emission tomography (PET) and the 5-HT1B specific radioligand [11C]AZ10419369 at baseline conditions. The binding potential (BPND) in cortical and subcortical areas was calculated using the simplified reference tissue model (SRTM). After correction for partial volume effects (PVEc), the correlation between age and regional BPND was examined. RESULTS A statistically significant negative correlation between age and BPND was obtained for neocortical regions and the ventral striatum (VST). The average reduction in BPND per decade was 8% in cortex and 4% in VST. The BPND in the caudate nucleus and the putamen was mainly unaffected by age. CONCLUSION The 5-HT1B receptor availability decreases by age in cortical regions, whereas it remains stable in the caudate nucleus and putamen. By consequence, age-matching of control subjects will be necessary in future clinical studies.
Collapse
Affiliation(s)
- Magdalena Nord
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden.
| | - Zsolt Cselenyi
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden; AstraZeneca Translational Science Center, Department of Clinical Neuroscience, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Anton Forsberg
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Göran Rosenqvist
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Mikael Tiger
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Johan Lundberg
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Andrea Varrone
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Lars Farde
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska University Hospital, SE-17176 Stockholm, Sweden; AstraZeneca Translational Science Center, Department of Clinical Neuroscience, Karolinska Institutet, SE-17176 Stockholm, Sweden
| |
Collapse
|
24
|
Tiger M, Rück C, Forsberg A, Varrone A, Lindefors N, Halldin C, Farde L, Lundberg J. Reduced 5-HT(1B) receptor binding in the dorsal brain stem after cognitive behavioural therapy of major depressive disorder. Psychiatry Res 2014; 223:164-70. [PMID: 24916155 DOI: 10.1016/j.pscychresns.2014.05.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 05/08/2014] [Accepted: 05/19/2014] [Indexed: 12/15/2022]
Abstract
Major depression is a significant contributor to the global burden of disease, and its pathophysiology is largely unknown. The serotonin hypothesis is, however, the model with most supporting data, although the details are only worked out to some extent. Recent clinical imaging measurements indeed imply a role in major depressive disorder (MDD) for the inhibitory serotonin autoreceptor 5-hydroxytryptamine1B (5-HT1B). The aim of the current study was to examine 5-HT1B receptor binding in the brain of MDD patients before and after psychotherapy. Ten patients with an ongoing untreated moderate depressive episode were examined with positron emission tomography (PET) and the 5-HT1B receptor selective radioligand [(11)C]AZ10419369, before and after treatment with internet-based cognitive behavioural therapy. All of the patients examined responded to treatment, and 70% were in remission by the time of the second PET measurement. A statistically significant 33% reduction of binding potential (BPND) was found in the dorsal brain stem (DBS) after treatment. No other significant changes in BPND were found. The DBS contains the raphe nuclei, which regulate the serotonin system. This study gives support for the importance of serotonin and the 5-HT1B receptor in the biological response to psychological treatment of MDD.
Collapse
Affiliation(s)
- Mikael Tiger
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden.
| | - Christian Rück
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Anton Forsberg
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Nils Lindefors
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Lars Farde
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| | - Johan Lundberg
- Department of Clinical Neuroscience, Karolinska Institutet, Centrum för psykiatriforskning, R5:0, Karolinska Universitetssjukhuset i Solna, 171 76 Stockholm, Sweden
| |
Collapse
|