1
|
Hart XM, Gründer G, Ansermot N, Conca A, Corruble E, Crettol S, Cumming P, Frajerman A, Hefner G, Howes O, Jukic MM, Kim E, Kim S, Maniscalco I, Moriguchi S, Müller DJ, Nakajima S, Osugo M, Paulzen M, Ruhe HG, Scherf-Clavel M, Schoretsanitis G, Serretti A, Spina E, Spigset O, Steimer W, Süzen SH, Uchida H, Unterecker S, Vandenberghe F, Verstuyft C, Zernig G, Hiemke C, Eap CB. Optimisation of pharmacotherapy in psychiatry through therapeutic drug monitoring, molecular brain imaging and pharmacogenetic tests: Focus on antipsychotics. World J Biol Psychiatry 2024; 25:451-536. [PMID: 38913780 DOI: 10.1080/15622975.2024.2366235] [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: 11/04/2023] [Revised: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND For psychotic disorders (i.e. schizophrenia), pharmacotherapy plays a key role in controlling acute and long-term symptoms. To find the optimal individual dose and dosage strategy, specialised tools are used. Three tools have been proven useful to personalise drug treatments: therapeutic drug monitoring (TDM) of drug levels, pharmacogenetic testing (PG), and molecular neuroimaging. METHODS In these Guidelines, we provide an in-depth review of pharmacokinetics, pharmacodynamics, and pharmacogenetics for 45 antipsychotics. Over 30 international experts in psychiatry selected studies that have measured drug concentrations in the blood (TDM), gene polymorphisms of enzymes involved in drug metabolism, or receptor/transporter occupancies in the brain (positron emission tomography (PET)). RESULTS Study results strongly support the use of TDM and the cytochrome P450 (CYP) genotyping and/or phenotyping to guide drug therapies. Evidence-based target ranges are available for titrating drug doses that are often supported by PET findings. CONCLUSION All three tools discussed in these Guidelines are essential for drug treatment. TDM goes well beyond typical indications such as unclear compliance and polypharmacy. Despite its enormous potential to optimise treatment effects, minimise side effects and ultimately reduce the global burden of diseases, personalised drug treatment has not yet become the standard of care in psychiatry.
Collapse
Affiliation(s)
- Xenia Marlene Hart
- Department of Molecular Neuroimaging, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Gerhard Gründer
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
- German Center for Mental Health (DZPG), Partner Site Mannheim, Heidelberg, Germany
| | - Nicolas Ansermot
- Department of Psychiatry, Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neuroscience, Lausanne University Hospital, Prilly, Switzerland
| | - Andreas Conca
- Dipartimento di Psichiatria, Comprensorio Sanitario di Bolzano, Bolzano, Italy
| | - Emmanuelle Corruble
- Service Hospitalo-Universitaire de Psychiatrie, Hôpital de Bicêtre, Université Paris-Saclay, AP-HP, Le Kremlin-Bicêtre, France
- Equipe MOODS, Inserm U1018, CESP (Centre de Recherche en Epidémiologie et Sante des Populations), Le Kremlin-Bicêtre, France
| | - Severine Crettol
- Department of Psychiatry, Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neuroscience, Lausanne University Hospital, Prilly, Switzerland
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
- School of Psychology and Counseling, Queensland University of Technology, Brisbane, Australia
| | - Ariel Frajerman
- Service Hospitalo-Universitaire de Psychiatrie, Hôpital de Bicêtre, Université Paris-Saclay, AP-HP, Le Kremlin-Bicêtre, France
- Equipe MOODS, Inserm U1018, CESP (Centre de Recherche en Epidémiologie et Sante des Populations), Le Kremlin-Bicêtre, France
| | - Gudrun Hefner
- Forensic Psychiatry, Vitos Clinic for Forensic Psychiatry, Eltville, Germany
| | - Oliver Howes
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, London, UK
| | - Marin M Jukic
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
- Pharmacogenetics Section, Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Euitae Kim
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seoyoung Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Ignazio Maniscalco
- Dipartimento di Psichiatria, Comprensorio Sanitario di Bolzano, Bolzano, Italy
| | - Sho Moriguchi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Daniel J Müller
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
- Pharmacogenetics Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Martin Osugo
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, London, UK
| | - Michael Paulzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- JARA - Translational Brain Medicine, Alexianer Center for Mental Health, Aachen, Germany
| | - Henricus Gerardus Ruhe
- Department of Psychiatry, Radboudumc, Nijmegen, Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| | - Maike Scherf-Clavel
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Georgios Schoretsanitis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | | | - Edoardo Spina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Olav Spigset
- Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Werner Steimer
- Institute of Clinical Chemistry and Pathobiochemistry, Technical University Munich, Munich, Germany
| | - Sinan H Süzen
- Department of Pharmaceutic Toxicology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Stefan Unterecker
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Frederik Vandenberghe
- Department of Psychiatry, Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neuroscience, Lausanne University Hospital, Prilly, Switzerland
| | - Celine Verstuyft
- Equipe MOODS, Inserm U1018, CESP (Centre de Recherche en Epidémiologie et Sante des Populations), Le Kremlin-Bicêtre, France
- Department of Molecular Genetics, Pharmacogenetics and Hormonology, Bicêtre University Hospital Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Gerald Zernig
- Department of Pharmacology, Medical University Innsbruck, Hall in Tirol, Austria
- Private Practice for Psychotherapy and Court-Certified Witness, Hall in Tirol, Austria
| | - Christoph Hiemke
- Department of Psychiatry and Psychotherapy and Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center of Mainz, Mainz, Germany
| | - Chin B Eap
- Department of Psychiatry, Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neuroscience, Lausanne University Hospital, Prilly, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Hart XM, Spangemacher M, Uchida H, Gründer G. Update Lessons from Positron Emission Tomography Imaging Part I: A Systematic Critical Review on Therapeutic Plasma Concentrations of Antipsychotics. Ther Drug Monit 2024; 46:16-32. [PMID: 38018857 DOI: 10.1097/ftd.0000000000001131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/06/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Positron emission tomography (PET) and single photon emission tomography (SPECT) of molecular drug targets (neuroreceptors and transporters) provide essential information for therapeutic drug monitoring-guided antipsychotic drug therapy. The optimal therapeutic windows for D 2 antagonists and partial agonists, as well as their proposed target ranges, are discussed based on an up-to-date literature search. METHODS This part I of II presents an overview of molecular neuroimaging studies in humans and primates involving the target engagement of amisulpride, haloperidol, clozapine, aripiprazole, olanzapine, quetiapine, risperidone, cariprazine, and ziprasidone. The systemic review particularly focused on dopamine D 2 -like and 5-HT 2A receptors. Target concentration ranges were estimated based on receptor occupancy ranges that relate to clinical effects or side effects (ie, extrapyramidal side effects). In addition, findings for other relevant receptor systems were included to further enrich the discussion. RESULTS The reported reference ranges for aripiprazole and clozapine align closely with findings from PET studies. Conversely, for haloperidol, risperidone, and olanzapine, the PET studies indicate that a lowering of the previously published upper limits would be necessary to decrease the risk of extrapyramidal side effect. CONCLUSIONS Molecular neuroimaging studies serve as a strong tool for defining target ranges for antipsychotic drug treatment and directing therapeutic drug monitoring.
Collapse
Affiliation(s)
- Xenia M Hart
- Central Institute of Mental Health, Department of Molecular Neuroimaging, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Moritz Spangemacher
- Central Institute of Mental Health, Department of Molecular Neuroimaging, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Central Institute of Mental Health, Department of Psychiatry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; and
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Gerhard Gründer
- Central Institute of Mental Health, Department of Molecular Neuroimaging, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
3
|
Li K, Liu X, Zhang M, Su R. Effects of Ketanserin, M100907 and Olanzapine on hallucinogenic like action induced by 2,5-dimethoxy-4-methylamphetamine. Behav Pharmacol 2023; 34:92-100. [PMID: 36752335 DOI: 10.1097/fbp.0000000000000693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
2,5-dimethoxy-4-methylamphetamine (DOM) is a kind of hallucinogen of phenylalkylamine. Psychedelic effects mainly include audiovisual synesthesia, complex imagery, disembodiment etc. that can impair control and cognition leading to adverse consequences such as suicide. By now, there are no specific drugs regarding the management of classic hallucinogen use clinically. We evaluated the effects of three 5-HT 2A receptor antagonists ketanseirn, M100907 and olanzapine on hallucination-like behavior in therapeutic and preventive administration with male C57BL/6J mice. Two models were used to evaluate the therapeutic potential of antagonists, one is head-twitch response (HTR) and the other is locomotion. Effects of ketanserin, M100907 and olanzapine on DOM-induced HTR were studied in preventive and therapeutic administration, respectively. In the preventive administration, the ID 50 values of ketanseirn, M100907 and olanzapine were 0.4 mg/kg, 0.005 mg/kg and 0.25 mg/kg. In the therapeutic administration, the ID 50 values of ketanseirn, M100907 and olanzapine were 0.04 mg/kg, 0.005 mg/kg and 0.03 mg/kg. Secondly, locomotor activity induced by DOM was performed to further evaluate the efficacy of three compounds. In locomotion, M100907(0.005 mg/kg) whenever in preventive or therapeutic administration, reduced the increase of movement distance induced by DOM. Although ketanserin (0.4 mg/kg) in the preventive administration also decreased the movement distance induced by DOM, it was alone administrated to influence the locomotor activity. Through HTR and locomotion, we compared the efficacy and latent side effects of ketanserin, M100907 and olanzapine against hallucinogenic like action induced by DOM. Our study provided additional experimental evidence on specific therapeutic drugs against hallucinogenic behavior induce by representative hallucinogen DOM.
Collapse
Affiliation(s)
- Kaixi Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing
- Shihezi University College of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, Department of Pharmacy, Xinjiang, China
| | - Xiaoyan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing
| | - Mei Zhang
- Shihezi University College of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi, Department of Pharmacy, Xinjiang, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing
| |
Collapse
|
4
|
Fu H, Rong J, Chen Z, Zhou J, Collier T, Liang SH. Positron Emission Tomography (PET) Imaging Tracers for Serotonin Receptors. J Med Chem 2022; 65:10755-10808. [PMID: 35939391 DOI: 10.1021/acs.jmedchem.2c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) and 5-HT receptors (5-HTRs) have crucial roles in various neuropsychiatric disorders and neurodegenerative diseases, making them attractive diagnostic and therapeutic targets. Positron emission tomography (PET) is a noninvasive nuclear molecular imaging technique and is an essential tool in clinical diagnosis and drug discovery. In this context, numerous PET ligands have been developed for "visualizing" 5-HTRs in the brain and translated into human use to study disease mechanisms and/or support drug development. Herein, we present a comprehensive repertoire of 5-HTR PET ligands by focusing on their chemotypes and performance in PET imaging studies. Furthermore, this Perspective summarizes recent 5-HTR-focused drug discovery, including biased agonists and allosteric modulators, which would stimulate the development of more potent and subtype-selective 5-HTR PET ligands and thus further our understanding of 5-HTR biology.
Collapse
Affiliation(s)
- Hualong Fu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Zhen Chen
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Jingyin Zhou
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Thomas Collier
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
5
|
Ebdrup BH, Knop FK. Weight gain on antipsychotics - A perfect storm of complex pathophysiology and psychopharmacology. Acta Psychiatr Scand 2021; 144:521-523. [PMID: 34587287 DOI: 10.1111/acps.13376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Bjørn H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research, CNSR & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
6
|
Peters EM, Bowen R, Balbuena L. Low-dose Quetiapine for Major Depressive Disorder and Sleep Improvement. J Clin Psychopharmacol 2021; 40:500-502. [PMID: 32796393 DOI: 10.1097/jcp.0000000000001262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Baltzersen OB, Meltzer HY, Frokjaer VG, Raghava JM, Baandrup L, Fagerlund B, Larsson HBW, Fibiger HC, Glenthøj BY, Knudsen GM, Ebdrup BH. Identification of a Serotonin 2A Receptor Subtype of Schizophrenia Spectrum Disorders With Pimavanserin: The Sub-Sero Proof-of-Concept Trial Protocol. Front Pharmacol 2020; 11:591. [PMID: 32425802 PMCID: PMC7204912 DOI: 10.3389/fphar.2020.00591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background All current approved antipsychotic drugs against schizophrenia spectrum disorders share affinity for the dopamine receptor (D2R). However, up to one-third of these patients respond insufficiently, and in some cases, side-effects outweigh symptom reduction. Previous data have suggested that a subgroup of antipsychotic-naïve patients will respond to serotonin 2A receptor (2AR) blockade. Aims This investigator-initiated, translational, proof-of-concept study has overall two aims; 1) To test the clinical effectiveness of monotherapy with the newly approved drug against Parkinson's disease psychosis, pimavanserin, in antipsychotic-free patients with first-episode schizophrenia spectrum disorders; 2) To characterize the neurobiological profile of responders to pimavaserin. Materials and Equipment Forty patients will be enrolled in this 6-week open label, one-armed trial with the selective serotonin 2AR antagonist (pimavanserin 34 mg/day). At baseline, patients will undergo: positron emission tomography (PET) imaging of the serotonin 2AR using the radioligand [¹¹C]Cimbi-36; structural magnetic resonance imaging (MRI); MR spectroscopy of cerebral glutamate levels and diffusion tensor imaging; cognitive and psychopathological examinations; electrocardiogram, and blood sampling for genetic- and metabolic analyses. Outcome Measures The primary clinical endpoint will be reduction in the Positive and Negative Syndrome Scale (PANSS) positive score. Secondary clinical endpoints comprise multiple clinical ratings (positive and negative symptoms, depressive-, obsessive-compulsive symptoms, quality of life, social functioning, sexual functioning, and side-effects). PET, MRI, and cognitive parameters will be used for in-depth neuropsychiatric characterization of pimavanserin response. Anticipated Results Clinically, we expect pimavanserin to reduce psychotic symptoms with similar effect as observed with conventional antipsychotics, for which we have comparable historical data. We expect pimavanserin to induce minimal side-effects. Neurobiologically, we expect psychotic symptom reduction to be most prominent in patients with low frontal serotonin 2AR binding potential at baseline. Potential pro-cognitive and brain structural effects of pimavanserin will be explored. Perspectives Sub-Sero will provide unique information about the role serotonin 2AR in antipsychotic-free, first-episode psychosis. If successful, Sub-Sero will aid identification of a “serotonergic subtype” of schizophrenia spectrum patients, thereby promoting development of precision medicine in clinical psychiatry. Clinical Trial Registration ClinicalTrials, identifier NCT03994965.
Collapse
Affiliation(s)
- Olga B Baltzersen
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention & Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, Glostrup, Denmark
| | - Herbert Y Meltzer
- Departments of Psychiatry and Behavioral Sciences, Pharmacology, and Physiology, School of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Vibe G Frokjaer
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Mental Health Services Copenhagen, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jayachandra M Raghava
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention & Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, Glostrup, Denmark.,Functional Imaging Unit (FIU), Rigshospitalet Glostrup, Glostrup, Denmark
| | - Lone Baandrup
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention & Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Fagerlund
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention & Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, Glostrup, Denmark
| | - Henrik B W Larsson
- Functional Imaging Unit (FIU), Rigshospitalet Glostrup, Glostrup, Denmark
| | - H Christian Fibiger
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Birte Y Glenthøj
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention & Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bjørn H Ebdrup
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention & Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
The 5-HT 6 receptor interactome: New insight in receptor signaling and its impact on brain physiology and pathologies. Neuropharmacology 2019; 172:107839. [PMID: 31682856 DOI: 10.1016/j.neuropharm.2019.107839] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 01/02/2023]
Abstract
The serotonin (5-HT)6 receptor is a Gs-coupled receptor exclusively expressed in the central nervous system. Highest receptor densities are found in brain regions implicated in mnemonic functions where the receptor is primarily but not exclusively located in the primary cilium of neurons. The 5-HT6 receptor continues to raise particular interest for neuropharmacologists, given the pro-cognitive effects of antagonists in a wide range of cognitive impairment paradigms in rodents and human. The 5-HT6 receptor also finely controls key neuro-developmental processes including neuron migration and differentiation. However, its influence upon neurodevelopment and cognition is not solely mediated by its coupling to the Gs-adenylyl cyclase pathway, suggesting alternative signal transduction mechanisms. This prompted studies aimed at characterizing the receptor interactome that identified 125 candidate receptor partners, making the 5-HT6 receptor one of the G protein-coupled receptors with the most extensively characterized interactome. These studies showed that the receptor localization at the plasma membrane and, consequently, its signal transduction, are finely modulated by several receptor partners. They demonstrated that prefrontal 5-HT6 receptors engage the mTOR pathway to compromise cognition in neurodevelopmental models of schizophrenia, and a role of the 5-HT6-mTOR pathway in temporal epilepsy. Finally, they revealed that the receptor activates Cdk5 signaling in an agonist-independent manner through a mechanism involving receptor phosphorylation by the associated Cdk5 and highlighted its key role in the migration of neurons and neurite growth. These new receptor-operated signaling mechanisms should be considered in the future development of drugs acting on 5-HT6 receptors. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
|
9
|
Azmanova M, Pitto-Barry A, Barry NPE. Schizophrenia: synthetic strategies and recent advances in drug design. MEDCHEMCOMM 2018; 9:759-782. [PMID: 30108966 PMCID: PMC6072500 DOI: 10.1039/c7md00448f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/09/2018] [Indexed: 12/19/2022]
Abstract
Schizophrenia is a complex and unpredictable mental disorder which affects several domains of cognition and behaviour. It is a heterogeneous illness characterised by positive, negative, and cognitive symptoms, often accompanied by signs of depression. In this tutorial review, we discuss recent progress in understanding the target sites and mechanisms of action of second-generation antipsychotic drugs. Progress in identifying and defining target sites has been accelerated recently by advances in neuroscience, and newly developed agents that regulate signalling by the main excitatory neurotransmitters in the brain are surveyed. Examples of novel molecules for the treatment of schizophrenia in preclinical and clinical development and their industrial sponsors are highlighted.
Collapse
Affiliation(s)
- Maria Azmanova
- School of Chemistry and Biosciences , University of Bradford , Bradford BD7 1DP , UK . ;
| | - Anaïs Pitto-Barry
- School of Chemistry and Biosciences , University of Bradford , Bradford BD7 1DP , UK . ;
| | - Nicolas P E Barry
- School of Chemistry and Biosciences , University of Bradford , Bradford BD7 1DP , UK . ;
| |
Collapse
|
10
|
Oranje B, Aggernaes B, Rasmussen H, Ebdrup BH, Glenthøj BY. Selective attention and mismatch negativity in antipsychotic-naïve, first-episode schizophrenia patients before and after 6 months of antipsychotic monotherapy. Psychol Med 2017; 47:2155-2165. [PMID: 28443529 DOI: 10.1017/s0033291717000599] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Attention deficits have been frequently reported in schizophrenia. It has been suggested that treatment with second-generation antipsychotics can ameliorate these deficits. In this study, the influence of 6 months treatment with quetiapine, a compound with less affinity for dopamine D2 receptors than for serotonergic 5-HT2A receptors, on electrophysiological parameters of attention was investigated in a group of antipsychotic-naïve, first-episode schizophrenia patients compared with a group of age- and gender-matched healthy controls. METHOD A total of 34 first-episode, antipsychotic-naïve patients with schizophrenia and an equal number of healthy controls were tested in a selective attention and a typical mismatch negativity (MMN) paradigm at baseline and after 6 months. The patients were treated with quetiapine according to their clinical needs during the period between baseline and follow-up, whereas controls received no treatment. RESULTS Patients showed lower MMN and P200 amplitude than healthy controls in the selective attention paradigm at baseline, while this was not the case for MMN of the typical MMN paradigm. Interestingly, after 6 months treatment, this MMN deficit was only ameliorated in patients treated with above median dosages of quetiapine. Patients had lower P3B amplitude, yet showed similar levels of processing negativity and N100 amplitude compared with healthy controls, both at baseline and follow-up. CONCLUSIONS The results indicate that deficits in MMN, P200 and P3B amplitude are present at early stages of schizophrenia, although depending on the paradigm used. Furthermore, the results indicate that 6 months quetiapine treatment ameliorates MMN but not P3B deficits, and only in those subjects on higher dosages.
Collapse
Affiliation(s)
- B Oranje
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS) and Center for Neuropsychiatric Schizophrenia Research (CNSR), Mental Health Services Glostrup,Capital Region Denmark, Glostrup,Denmark
| | - B Aggernaes
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS) and Center for Neuropsychiatric Schizophrenia Research (CNSR), Mental Health Services Glostrup,Capital Region Denmark, Glostrup,Denmark
| | - H Rasmussen
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS) and Center for Neuropsychiatric Schizophrenia Research (CNSR), Mental Health Services Glostrup,Capital Region Denmark, Glostrup,Denmark
| | - B H Ebdrup
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS) and Center for Neuropsychiatric Schizophrenia Research (CNSR), Mental Health Services Glostrup,Capital Region Denmark, Glostrup,Denmark
| | - B Y Glenthøj
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS) and Center for Neuropsychiatric Schizophrenia Research (CNSR), Mental Health Services Glostrup,Capital Region Denmark, Glostrup,Denmark
| |
Collapse
|
11
|
Michels L, Scherpiet S, Stämpfli P, Herwig U, Brühl AB. Baseline Perfusion Alterations Due to Acute Application of Quetiapine and Pramipexole in Healthy Adults. Int J Neuropsychopharmacol 2016; 19:pyw067. [PMID: 27466220 PMCID: PMC5137281 DOI: 10.1093/ijnp/pyw067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 07/01/2016] [Accepted: 07/22/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The dopaminergic system is implicated in many mental processes and neuropsychiatric disorders. Pharmacologically, drugs with dopamine receptor antagonistic and agonistic effects are used, but their effects on functional brain metabolism are not well known. METHODS In this randomized crossover, placebo-controlled, and rater-blinded study, 25 healthy adults received an acute dose placebo substance (starch), quetiapine (dopamine receptor antagonist), or pramipexole (dopamine agonist of the nonergoline class) 1 hour before the experiment. Background-suppressed 2D pseudo-continuous arterial spin labeling was used to examine whole-brain baseline cerebral blood flow differences induced by the 3 substances. RESULTS We found that quetiapine reduced perfusion in the occipital (early visual areas) and bilateral cerebellar cortex relative to placebo. In contrast, quetiapine enhanced cerebral blood flow (relative to placebo) in the striatal system (putamen and caudate nucleus) but also in the supplementary motor area, insular-, prefrontal- as well as in the pre- and postcentral cortex. Pramipexole increased cerebral blood flow compared with placebo in the caudate nucleus, putamen, middle frontal, supplementary motor area, and brainstem (substantia nigra), but reduced cerebral blood flow in the posterior thalamus, cerebellum, and visual areas. Pramipexole administration resulted in stronger cerebral blood flow relative to quetiapine in the hypothalamus, cerebellum, and substantia nigra. CONCLUSIONS Our results indicate that quetiapine and pramipexole differentially modulate regional baseline cerebral blood flow. Both substances act on the dopaminergic system, although they affect distinct regions. Quetiapine altered dopaminergic function in frontal, striatal, and motor regions. In contrast, pramipexole affected cerebral blood flow of the nigrostriatal (striatum and substantia nigra) dopaminergic, but less the fronto-insular system.
Collapse
Affiliation(s)
- Lars Michels
- Institute of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (Dr Michels); MR-Center, University Children's Hospital Zurich, Zurich, Switzerland (Dr Michels); Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland (Drs Scherpiet, Stämpfli, Herwig, and Brühl); Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK (Dr Brühl). .,L.M. and S.S. are shared first authors.
| | | | | | | | | |
Collapse
|
12
|
Abstract
BACKGROUND A significant number of patients with schizophrenia fail to respond to antipsychotic medication. Although several studies have investigated associated patient characteristics, the emerging findings from genetic studies offer further scope for study. METHOD In 612 schizophrenia patients with detailed clinical information, common genetic variants indexed by polygenic risk scores, and rare variants indexed by deletion and duplication burden genomewide, we explored potential genetic predictors alongside other established risk factors for treatment resistance. Clinical outcomes of treatment resistance were also calculated using lifetime measures of positive, negative/disorganized and mood symptoms as well as number of hospitalizations and suicide attempts. RESULTS Logistic regression models identified a significant relationship between treatment resistance and total duplication burden genomewide, years of formal schooling and age at onset. Clinically, treatment-resistant patients were characterized by greater negative/disorganized and positive symptoms and greater number of hospitalizations. CONCLUSIONS Taken together, these findings suggest genetic information, specifically the genomewide burden of rare copy number variants, may increase our understanding and clinical management of patients with treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- A K Martin
- Queensland Brain Institute,University of Queensland,St Lucia,Brisbane 4072,Australia
| | - B Mowry
- Queensland Brain Institute,University of Queensland,St Lucia,Brisbane 4072,Australia
| |
Collapse
|
13
|
Nørbak-Emig H, Ebdrup BH, Fagerlund B, Svarer C, Rasmussen H, Friberg L, Allerup PN, Rostrup E, Pinborg LH, Glenthøj BY. Frontal D2/3 Receptor Availability in Schizophrenia Patients Before and After Their First Antipsychotic Treatment: Relation to Cognitive Functions and Psychopathology. Int J Neuropsychopharmacol 2016; 19:pyw006. [PMID: 26819282 PMCID: PMC4886673 DOI: 10.1093/ijnp/pyw006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/18/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND We have previously reported associations between frontal D2/3 receptor binding potential positive symptoms and cognitive deficits in antipsychotic-naïve schizophrenia patients. Here, we examined the effect of dopamine D2/3 receptor blockade on cognition. Additionally, we explored the relation between frontal D2/3 receptor availability and treatment effect on positive symptoms. METHODS Twenty-five antipsychotic-naïve first-episode schizophrenia patients were examined with the Positive and Negative Syndrome Scale, tested with the cognitive test battery Cambridge Neuropsychological Test Automated Battery, scanned with single-photon emission computerized tomography using the dopamine D2/3 receptor ligand [(123)I]epidepride, and scanned with MRI. After 3 months of treatment with either risperidone (n=13) or zuclopenthixol (n=9), 22 patients were reexamined. RESULTS Blockade of extrastriatal dopamine D2/3 receptors was correlated with decreased attentional focus (r = -0.615, P=.003) and planning time (r = -0.436, P=.048). Moreover, baseline frontal dopamine D2/3 binding potential and positive symptom reduction correlated positively (D2/3 receptor binding potential left frontal cortex rho = 0.56, P=.003; D2/3 receptor binding potential right frontal cortex rho = 0.48, P=.016). CONCLUSIONS Our data support the hypothesis of a negative influence of D2/3 receptor blockade on specific cognitive functions in schizophrenia. This is highly clinically relevant given the well-established association between severity of cognitive disturbances and a poor functional outcome in schizophrenia. Additionally, the findings support associations between frontal D2/3 receptor binding potential at baseline and the effect of antipsychotic treatment on positive symptoms.
Collapse
Affiliation(s)
- Henrik Nørbak-Emig
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Bjørn H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Birgitte Fagerlund
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Claus Svarer
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Hans Rasmussen
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Lars Friberg
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Peter N Allerup
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Egill Rostrup
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Lars H Pinborg
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg)
| | - Birte Y Glenthøj
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Mental Health Centre Glostrup, Mental Health Services, Capital Region of Denmark, Glostrup, Denmark (Drs Nørbak-Emig, Ebdrup, Fagerlund, Rasmussen, and Glenthøj); University of Copenhagen, Faculty of Health and Medical Sciences, Department of Clinical Medicine, Copenhagen, Denmark (Drs Nørbak-Emig and Glenthøj); Neurobiology Research Unit and Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark (Drs Svarer and Pinborg); Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark (Dr Allerup); Functional Imaging Unit, Department of Diagnostics, Copenhagen University Hospital, Glostrup Hospital, Denmark (Dr Rostrup); Department of Clinical Physiology and Nuclear Medicine, Copenhagen University, Bispebjerg Hospital, Denmark (Dr Friberg).
| |
Collapse
|
14
|
Wulff S, Pinborg LH, Svarer C, Jensen LT, Nielsen MØ, Allerup P, Bak N, Rasmussen H, Frandsen E, Rostrup E, Glenthøj BY. Striatal D(2/3) Binding Potential Values in Drug-Naïve First-Episode Schizophrenia Patients Correlate With Treatment Outcome. Schizophr Bull 2015; 41:1143-52. [PMID: 25698711 PMCID: PMC4535636 DOI: 10.1093/schbul/sbu220] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
One of best validated findings in schizophrenia research is the association between blockade of dopamine D2 receptors and the effects of antipsychotics on positive psychotic symptoms. The aim of the present study was to examine correlations between baseline striatal D(2/3) receptor binding potential (BP(p)) values and treatment outcome in a cohort of antipsychotic-naïve first-episode schizophrenia patients. Additionally, we wished to investigate associations between striatal dopamine D(2/3) receptor blockade and alterations of negative symptoms as well as functioning and subjective well-being. Twenty-eight antipsychotic-naïve schizophrenia patients and 26 controls were included in the study. Single-photon emission computed tomography (SPECT) with [(123)I]iodobenzamide ([(123)I]-IBZM) was used to examine striatal D(2/3) receptor BP(p). Patients were examined before and after 6 weeks of treatment with the D(2/3) receptor antagonist amisulpride. There was a significant negative correlation between striatal D(2/3) receptor BP(p) at baseline and improvement of positive symptoms in the total group of patients. Comparing patients responding to treatment to nonresponders further showed significantly lower baseline BP(p) in the responders. At follow-up, the patients demonstrated a negative correlation between the blockade and functioning, whereas no associations between blockade and negative symptoms or subjective well-being were observed. The results show an association between striatal BP(p) of dopamine D(2/3) receptors in antipsychotic-naïve first-episode patients with schizophrenia and treatment response. Patients with a low BP(p) have a better treatment response than patients with a high BP(p). The results further suggest that functioning may decline at high levels of dopamine receptor blockade.
Collapse
Affiliation(s)
- Sanne Wulff
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Psychiatric Center Glostrup, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Denmark;
| | - Lars Hageman Pinborg
- Neurobiology Research Unit (NRU), Rigshospitalet, University of Copenhagen, Denmark
| | - Claus Svarer
- Neurobiology Research Unit (NRU), Rigshospitalet, University of Copenhagen, Denmark
| | - Lars Thorbjørn Jensen
- Department of Clinical Physiology and Nuclear Medicine, Herlev Hospital, University of Copenhagen, Denmark
| | - Mette Ødegaard Nielsen
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and,Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Psychiatric Center Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Peter Allerup
- Department of Education, Centre for Research in Compulsory Schooling, Aarhus University, Denmark
| | - Nikolaj Bak
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and,Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Psychiatric Center Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Hans Rasmussen
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and,Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Psychiatric Center Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Erik Frandsen
- Department of Diagnostics, Functional Imaging Unit and Section of Clinical Physiology and Nuclear Medicine, Glostrup Hospital, University of Copenhagen, Denmark
| | - Egill Rostrup
- Department of Diagnostics, Functional Imaging Unit and Section of Clinical Physiology and Nuclear Medicine, Glostrup Hospital, University of Copenhagen, Denmark
| | - Birte Yding Glenthøj
- Center for Neuropsychiatric Schizophrenia Research (CNSR) and,Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Psychiatric Center Glostrup, University of Copenhagen, Copenhagen, Denmark;,Department of Clinical Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Denmark
| |
Collapse
|
15
|
Knudsen GM, Jensen PS, Erritzoe D, Baaré WFC, Ettrup A, Fisher PM, Gillings N, Hansen HD, Hansen LK, Hasselbalch SG, Henningsson S, Herth MM, Holst KK, Iversen P, Kessing LV, Macoveanu J, Madsen KS, Mortensen EL, Nielsen FÅ, Paulson OB, Siebner HR, Stenbæk DS, Svarer C, Jernigan TL, Strother SC, Frokjaer VG. The Center for Integrated Molecular Brain Imaging (Cimbi) database. Neuroimage 2015; 124:1213-1219. [PMID: 25891375 DOI: 10.1016/j.neuroimage.2015.04.025] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 01/07/2023] Open
Abstract
We here describe a multimodality neuroimaging containing data from healthy volunteers and patients, acquired within the Lundbeck Foundation Center for Integrated Molecular Brain Imaging (Cimbi) in Copenhagen, Denmark. The data is of particular relevance for neurobiological research questions related to the serotonergic transmitter system with its normative data on the serotonergic subtype receptors 5-HT1A, 5-HT1B, 5-HT2A, and 5-HT4 and the 5-HT transporter (5-HTT), but can easily serve other purposes. The Cimbi database and Cimbi biobank were formally established in 2008 with the purpose to store the wealth of Cimbi-acquired data in a highly structured and standardized manner in accordance with the regulations issued by the Danish Data Protection Agency as well as to provide a quality-controlled resource for future hypothesis-generating and hypothesis-driven studies. The Cimbi database currently comprises a total of 1100 PET and 1000 structural and functional MRI scans and it holds a multitude of additional data, such as genetic and biochemical data, and scores from 17 self-reported questionnaires and from 11 neuropsychological paper/computer tests. The database associated Cimbi biobank currently contains blood and in some instances saliva samples from about 500 healthy volunteers and 300 patients with e.g., major depression, dementia, substance abuse, obesity, and impulsive aggression. Data continue to be added to the Cimbi database and biobank.
Collapse
Affiliation(s)
- Gitte M Knudsen
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark.
| | - Peter S Jensen
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - David Erritzoe
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - William F C Baaré
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - Anders Ettrup
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Patrick M Fisher
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Nic Gillings
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; PET and Cyclotron Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Hanne D Hansen
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Lars Kai Hansen
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; DTU Compute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Steen G Hasselbalch
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Susanne Henningsson
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - Matthias M Herth
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; PET and Cyclotron Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Klaus K Holst
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Department of Biostatistics, University of Copenhagen, DK-1014 Copenhagen, Denmark
| | - Pernille Iversen
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - Lars V Kessing
- Psychiatric Center Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Julian Macoveanu
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark; Psychiatric Center Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kathrine Skak Madsen
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark
| | - Erik L Mortensen
- Department of Public Health and Center for Healthy Aging, University of Copenhagen, DK-2200 Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Finn Årup Nielsen
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; DTU Compute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Olaf B Paulson
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Hartwig R Siebner
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, DK-2650 Hvidovre, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg, DK-2400 Copenhagen, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg, DK-2400 Copenhagen, Denmark
| | - Dea S Stenbæk
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Claus Svarer
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Terry L Jernigan
- Center for Human Development, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephen C Strother
- Rotman Research Institute, Baycrest Centre, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Vibe G Frokjaer
- Center for Integrated Molecular Brain Imaging, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark; Psychiatric Center Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
16
|
Neocortical serotonin2A receptor binding predicts quetiapine associated weight gain in antipsychotic-naive first-episode schizophrenia patients. Int J Neuropsychopharmacol 2014; 17:1729-36. [PMID: 24830305 DOI: 10.1017/s1461145714000777] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Antipsychotic-induced weight gain is of major clinical importance since it is associated with severe metabolic complications and increased mortality. The serotonin2A receptor system has been suggested to be implicated in weight gain and obesity. However, no previous in vivo imaging data have related serotonin2A receptor binding to weight gain before and after antipsychotic monotherapy. Fifteen antipsychotic-naive first-episode schizophrenia patients were included and investigated before and after six months of quetiapine treatment. We examined the relationship between serotonin2A receptor binding as measured with positron emission tomography (PET) and [18F]altanserin and change in body mass index (BMI). Quetiapine was chosen because it is characterized by a moderately high affinity for the serotonin2A receptor and a fast dissociation rate from the dopamine D2 receptor. At baseline the mean BMI was 24.2 kg/m2, range 18-36 kg/m2. After six months of quetiapine treatment (mean dose: 383 mg/day) the BMI had, on average, increased by 6.7%, corresponding to an average weight gain of 5.0 kg. We found a significant positive correlation both between neocortical serotonin2A receptor binding prior to treatment and subsequent increase in BMI (rho=0.59, p=0.022). At follow-up, the serotonin2A receptor occupancy was positively correlated with BMI increase (rho=0.54, p=0.038). To our knowledge, these are the first in vivo receptor imaging data in initially antipsychotic-naive first-episode schizophrenia patients to show that the cerebral serotonin2A receptor is associated with antipsychotic-induced weight gain.
Collapse
|
17
|
Ren H, Wey HY, Strebl M, Neelamegam R, Ritter T, Hooker JM. Synthesis and imaging validation of [¹⁸F]MDL100907 enabled by Ni-mediated fluorination. ACS Chem Neurosci 2014; 5:611-5. [PMID: 24845956 DOI: 10.1021/cn500078e] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Several voids exist in reliable positron emission tomography (PET) radioligands for quantification of the serotonin (5HT) receptor system. Even in cases where 5HT radiotracers exist, challenges remain that have limited the utility of 5HT imaging in clinical research. Herein we address an unmet need in 5HT2a imaging using innovative chemistry. We report a scalable and robust synthesis of [(18)F]MDL100907, which was enabled by a Ni-mediated oxidative fluorination using [(18)F]fluoride. This first demonstration of a Ni-mediated fluorination used for PET imaging required development of a new reaction strategy that ultimately provided high specific activity [(18)F]MDL100907. Using the new synthetic strategy and optimized procedure, [(18)F]MDL100907 was evaluated against [(11)C]MDL100907 for reliability to quantify 5HT₂a in the nonhuman primate brain and was found to be superior based on a single scan analysis using the same nonhuman primate. The use of this new 5HT₂a radiotracer will afford clinical neuroscience research the ability to distinguish 5HT₂a receptor abnormalities binding between healthy subjects and patients even when group differences are small.
Collapse
Affiliation(s)
- Hong Ren
- Athinoula A. Martinos
Center for Biomedical Imaging, Massachusetts General Hospital and
Harvard Medical School, 13th Street, Charlestown, Massachusetts 02129, United States
- Department
of Chemistry and Chemical Biology, Harvard University, 12 Oxford
Street, Cambridge, Massachusetts 02138, United States
| | - Hsiao-Ying Wey
- Athinoula A. Martinos
Center for Biomedical Imaging, Massachusetts General Hospital and
Harvard Medical School, 13th Street, Charlestown, Massachusetts 02129, United States
| | - Martin Strebl
- Department
of Chemistry and Chemical Biology, Harvard University, 12 Oxford
Street, Cambridge, Massachusetts 02138, United States
| | - Ramesh Neelamegam
- Athinoula A. Martinos
Center for Biomedical Imaging, Massachusetts General Hospital and
Harvard Medical School, 13th Street, Charlestown, Massachusetts 02129, United States
| | - Tobias Ritter
- Department
of Chemistry and Chemical Biology, Harvard University, 12 Oxford
Street, Cambridge, Massachusetts 02138, United States
| | - Jacob M. Hooker
- Athinoula A. Martinos
Center for Biomedical Imaging, Massachusetts General Hospital and
Harvard Medical School, 13th Street, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
18
|
Serotonin 2A receptor agonist binding in the human brain with [¹¹C]Cimbi-36. J Cereb Blood Flow Metab 2014; 34:1188-96. [PMID: 24780897 PMCID: PMC4083382 DOI: 10.1038/jcbfm.2014.68] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 11/08/2022]
Abstract
[(11)C]Cimbi-36 was recently developed as a selective serotonin 2A (5-HT(2A)) receptor agonist radioligand for positron emission tomography (PET) brain imaging. Such an agonist PET radioligand may provide a novel, and more functional, measure of the serotonergic system and agonist binding is more likely than antagonist binding to reflect 5-HT levels in vivo. Here, we show data from a first-in-human clinical trial with [(11)C]Cimbi-36. In 29 healthy volunteers, we found high brain uptake and distribution according to 5-HT(2A) receptors with [(11)C]Cimbi-36 PET. The two-tissue compartment model using arterial input measurements provided the most optimal quantification of cerebral [(11)C]Cimbi-36 binding. Reference tissue modeling was feasible as it induced a negative but predictable bias in [(11)C]Cimbi-36 PET outcome measures. In five subjects, pretreatment with the 5-HT(2A) receptor antagonist ketanserin before a second PET scan significantly decreased [(11)C]Cimbi-36 binding in all cortical regions with no effects in cerebellum. These results confirm that [(11)C]Cimbi-36 binding is selective for 5-HT(2A) receptors in the cerebral cortex and that cerebellum is an appropriate reference tissue for quantification of 5-HT(2A) receptors in the human brain. Thus, we here describe [(11)C]Cimbi-36 as the first agonist PET radioligand to successfully image and quantify 5-HT(2A) receptors in the human brain.
Collapse
|
19
|
Manganese-enhanced magnetic resonance imaging reveals increased DOI-induced brain activity in a mouse model of schizophrenia. Proc Natl Acad Sci U S A 2014; 111:E2492-500. [PMID: 24889602 DOI: 10.1073/pnas.1323287111] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Maternal infection during pregnancy increases the risk for schizophrenia in offspring. In rodent models, maternal immune activation (MIA) yields offspring with schizophrenia-like behaviors. None of these behaviors are, however, specific to schizophrenia. The presence of hallucinations is a key diagnostic symptom of schizophrenia. In mice, this symptom can be defined as brain activation in the absence of external stimuli, which can be mimicked by administration of hallucinogens. We find that, compared with controls, adult MIA offspring display an increased stereotypical behavioral response to the hallucinogen 2,5-dimethoxy-4-iodoamphetamine (DOI), an agonist for serotonin receptor 2A (5-HT2AR). This may be explained by increased levels of 5-HT2AR and downstream signaling molecules in unstimulated MIA prefrontal cortex (PFC). Using manganese-enhanced magnetic resonance imaging to identify neuronal activation elicited by DOI administration, we find that, compared with controls, MIA offspring exhibit a greater manganese (Mn(2+)) accumulation in several brain areas, including the PFC, thalamus, and striatum. The parafascicular thalamic nucleus, which plays the role in the pathogenesis of hallucinations, is activated by DOI in MIA offspring only. Additionally, compared with controls, MIA offspring demonstrate higher DOI-induced expression of early growth response protein 1, cyclooxygenase-2, and brain-derived neurotrophic factor in the PFC. Chronic treatment with the 5-HT2AR antagonist ketanserin reduces DOI-induced head twitching in MIA offspring. Thus, the MIA mouse model can be successfully used to investigate activity induced by DOI in awake, behaving mice. Moreover, manganese-enhanced magnetic resonance imaging is a useful, noninvasive method for accurately measuring this type of activity.
Collapse
|
20
|
Ebdrup BH, Nørbak H, Borgwardt S, Glenthøj B. Volumetric changes in the basal ganglia after antipsychotic monotherapy: a systematic review. Curr Med Chem 2014; 20:438-47. [PMID: 23157636 PMCID: PMC3715891 DOI: 10.2174/0929867311320030015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 09/09/2012] [Accepted: 09/26/2012] [Indexed: 11/22/2022]
Abstract
Introduction: Exposure to antipsychotic medication has been extensively associated with structural brain changes in the basal ganglia (BG). Traditionally antipsychotics have been divided into first and second generation antipsychotics (FGAs and SGAs) however, the validity of this classification has become increasingly controversial. To address if specific antipsychotics induce differential effects on BG volumes or whether volumetric effects are explained by FGA or SGA classification, we reviewed longitudinal structural magnetic resonance imaging (MRI) studies investigating effects of antipsychotic monotherapy. Material and Methods: We systematically searched PubMed for longitudinal MRI studies of patients with schizophrenia or non-affective psychosis who had undergone a period of antipsychotic monotherapy. We used specific, predefined search terms and extracted studies were hand searched for additional studies. Results: We identified 13 studies published in the period from 1996 to 2011. Overall six compounds (two classified as FGAs and four as SGAs) have been investigated: haloperidol, zuclophentixol, risperidone, olanzapine, clozapine, and quetiapine. The follow-up period ranged from 3-24 months. Unexpectedly, no studies found that specific FGAs induce significant BG volume increases. Conversely, both volumetric increases and decreases in the BG have been associated with SGA monotherapy. Discussion: Induction of striatal volume increases is not a specific feature of FGAs. Except for clozapine treatment in chronic patients, volume reductions are not restricted to specific SGAs. The current review adds brain structural support to the notion that antipsychotics should no longer be classified as either FGAs or SGAs. Future clinical MRI studies should strive to elucidate effects of specific antipsychotic drugs.
Collapse
Affiliation(s)
- B H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research, CNSR & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, Psychiatric Center Glostrup, University Hospital DK-Glostrup, Denmark.
| | | | | | | |
Collapse
|
21
|
Zimmer L, Le Bars D. Current status of positron emission tomography radiotracers for serotonin receptors in humans. J Labelled Comp Radiopharm 2013; 56:105-13. [DOI: 10.1002/jlcr.3001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 11/08/2012] [Accepted: 11/08/2012] [Indexed: 12/19/2022]
|
22
|
Norquetiapine and depressive symptoms in initially antipsychotic-naive first-episode schizophrenia. J Clin Psychopharmacol 2013; 33:266-9. [PMID: 23422401 DOI: 10.1097/jcp.0b013e318287acc9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
23
|
Mestre TA, Zurowski M, Fox SH. 5-Hydroxytryptamine 2A receptor antagonists as potential treatment for psychiatric disorders. Expert Opin Investig Drugs 2013; 22:411-21. [DOI: 10.1517/13543784.2013.769957] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
24
|
Halberstadt AL, Powell SB, Geyer MA. Role of the 5-HT₂A receptor in the locomotor hyperactivity produced by phenylalkylamine hallucinogens in mice. Neuropharmacology 2013; 70:218-27. [PMID: 23376711 DOI: 10.1016/j.neuropharm.2013.01.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 12/20/2012] [Accepted: 01/12/2013] [Indexed: 01/12/2023]
Abstract
The 5-HT₂A receptor mediates the effects of serotonergic hallucinogens and may play a role in the pathophysiology of certain psychiatric disorders, including schizophrenia. Given these findings, there is a need for animal models to assess the behavioral effects of 5-HT₂A receptor activation. Our previous studies demonstrated that the phenylalkylamine hallucinogen and 5-HT₂A/₂C agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) produces dose-dependent effects on locomotor activity in C57BL/6J mice, increasing activity at low to moderate doses and reducing activity at high doses. DOI did not increase locomotor activity in 5-HT₂A knockout mice, indicating the effect is a consequence of 5-HT₂A receptor activation. Here, we tested a series of phenylalkylamine hallucinogens in C57BL/6J mice using the Behavioral Pattern Monitor (BPM) to determine whether these compounds increase locomotor activity by activating the 5-HT₂A receptor. Low doses of mescaline, 2,5-dimethoxy-4-ethylamphetamine (DOET), 2,5-dimethoxy-4-propylamphetamine (DOPR), 2,4,5-trimethoxyamphetamine (TMA-2), and the conformationally restricted phenethylamine (4-bromo-3,6-dimethoxybenzocyclobuten-1-yl)methylamine (TCB-2) increased locomotor activity. By contrast, the non-hallucinogenic phenylalkylamine 2,5-dimethoxy-4-tert-butylamphetamine (DOTB) did not alter locomotor activity at any dose tested (0.1-10 mg/kg i.p.). The selective 5-HT₂A antagonist M100907 blocked the locomotor hyperactivity induced by mescaline and TCB-2. Similarly, mescaline and TCB-2 did not increase locomotor activity in 5-HT₂A knockout mice. These results confirm that phenylalkylamine hallucinogens increase locomotor activity in mice and demonstrate that this effect is mediated by 5-HT₂A receptor activation. Thus, locomotor hyperactivity in mice can be used to assess phenylalkylamines for 5-HT₂A agonist activity and hallucinogen-like behavioral effects. These studies provide additional support for the link between 5-HT₂A activation and hallucinogenesis.
Collapse
Affiliation(s)
- Adam L Halberstadt
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0804, USA.
| | | | | |
Collapse
|
25
|
Hammer TB, Oranje B, Skimminge A, Aggernæs B, Ebdrup BH, Glenthøj B, Baaré W. Structural brain correlates of sensorimotor gating in antipsychotic-naive men with first-episode schizophrenia. J Psychiatry Neurosci 2013; 38:34-42. [PMID: 22687247 PMCID: PMC3529217 DOI: 10.1503/jpn.110129] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Prepulse inhibition (PPI) of the startle reflex is modulated by a complex neural network. Prepulse inhibition impairments are found at all stages of schizophrenia. Previous magnetic resonance imaging (MRI) studies suggest that brain correlates of PPI differ between patients with schizophrenia and healthy controls; however, these studies included only patients with chronic illness and medicated patients. Our aim was to examine the structural brain correlates of PPI in antipsychotic-naive patients with first-episode schizophrenia. METHODS We performed acoustic PPI assessment and structural MRI (1.5 and 3 T) in men with first-episode schizophrenia and age-matched controls. Voxel-based morphometry was used to investigate the association between PPI and grey matter volumes. RESULTS We included 27 patients and 38 controls in the study. Patients had lower PPI than controls. The brain areas in which PPI and grey matter volume correlated did not differ between the groups. Independent of group, PPI was significantly and positively associated with regional grey matter volume in the right superior parietal cortex. Prepulse inhibition and grey matter volume associations were also observed in the left rostral dorsal premotor cortex, the right presupplementary motor area and the anterior medial superior frontal gyrus bilaterally. Follow-up analyses suggested that the rostral dorsal premotor cortex and presupplementary motor area correlations were driven predominantly by the controls. LIMITATIONS We used 2 different MRI scanners, which might have limited our ability to find subcortical associations since interscanner consistency is low for subcortical regions. CONCLUSION The superior parietal cortex seems to be involved in the regulation of PPI in controls and antipsychotic-naive men with first-episode schizophrenia. Our observation that PPI deficits in schizophrenia may be related to the rostral dorsal premotor cortex and presupplementary motor area, brain areas involved in maintaining relevant sensory information and voluntary inhibition, warrants further study.
Collapse
Affiliation(s)
- Trine Bjørg Hammer
- Center for Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital, Glostrup, Denmark.
| | | | | | | | | | | | | |
Collapse
|
26
|
Risgaard R, Ettrup A, Balle T, Dyssegaard A, Hansen HD, Lehel S, Madsen J, Pedersen H, Püschl A, Badolo L, Bang-Andersen B, Knudsen GM, Kristensen JL. Radiolabelling and PET brain imaging of the α1-adrenoceptor antagonist Lu AE43936. Nucl Med Biol 2013; 40:135-40. [DOI: 10.1016/j.nucmedbio.2012.09.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/10/2012] [Accepted: 09/27/2012] [Indexed: 10/27/2022]
|
27
|
Roberts PD, Spiros A, Geerts H. Simulations of symptomatic treatments for Alzheimer's disease: computational analysis of pathology and mechanisms of drug action. ALZHEIMERS RESEARCH & THERAPY 2012. [PMID: 23181523 PMCID: PMC3580459 DOI: 10.1186/alzrt153] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction A substantial number of therapeutic drugs for Alzheimer's disease (AD) have failed in late-stage trials, highlighting the translational disconnect with pathology-based animal models. Methods To bridge the gap between preclinical animal models and clinical outcomes, we implemented a conductance-based computational model of cortical circuitry to simulate working memory as a measure for cognitive function. The model was initially calibrated using preclinical data on receptor pharmacology of catecholamine and cholinergic neurotransmitters. The pathology of AD was subsequently implemented as synaptic and neuronal loss and a decrease in cholinergic tone. The model was further calibrated with clinical Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-Cog) results on acetylcholinesterase inhibitors and 5-HT6 antagonists to improve the model's prediction of clinical outcomes. Results As an independent validation, we reproduced clinical data for apolipoprotein E (APOE) genotypes showing that the ApoE4 genotype reduces the network performance much more in mild cognitive impairment conditions than at later stages of AD pathology. We then demonstrated the differential effect of memantine, an N-Methyl-D-aspartic acid (NMDA) subunit selective weak inhibitor, in early and late AD pathology, and show that inhibition of the NMDA receptor NR2C/NR2D subunits located on inhibitory interneurons compensates for the greater excitatory decline observed with pathology. Conclusions This quantitative systems pharmacology approach is shown to be complementary to traditional animal models, with the potential to assess potential off-target effects, the consequences of pharmacologically active human metabolites, the effect of comedications, and the impact of a small number of well described genotypes.
Collapse
Affiliation(s)
- Patrick D Roberts
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue, Portland, OR 97239 USA ; In Silico Biosciences, Inc., 405 Waltham Street, Lexington, MA 02421 USA
| | - Athan Spiros
- In Silico Biosciences, Inc., 405 Waltham Street, Lexington, MA 02421 USA
| | - Hugo Geerts
- In Silico Biosciences, Inc., 405 Waltham Street, Lexington, MA 02421 USA
| |
Collapse
|
28
|
Schennach R, Riedel M, Musil R, Möller HJ. Treatment Response in First-episode Schizophrenia. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2012; 10:78-87. [PMID: 23430971 PMCID: PMC3569147 DOI: 10.9758/cpn.2012.10.2.78] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 05/31/2012] [Indexed: 12/18/2022]
Abstract
First episode schizophrenia (FES) patients tend to be more responsive to treatment. An adequate response has been associated with a favourable long-term course in FES patients. Yet, despite the generally very favourable response profile around one quarter of the patients shows persisting symptoms of psychosis. To improve the outcome and course of psychosis great effort has emerged in identifying biological and clinical variables associated with non-response in order to identify non-responders as early as possible and adopt specific treatment strategies improving illness outcome. Different antipsychotic treatment regimens have been evaluated in terms of their efficacy in reducing symptoms of FES with psychological interventions gaining increasing importance in the treatment concept of patients suffering from their first illness episode. Therefore, aim of this review is to summarize current evidence on the response patterns, the most important predictors of response/non-response as well as on effective treatment interventions in FES patients.
Collapse
Affiliation(s)
- Rebecca Schennach
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | | |
Collapse
|
29
|
Plasma Concentrations of Quetiapine, N-Desalkylquetiapine, O-Desalkylquetiapine, 7-Hydroxyquetiapine, and Quetiapine Sulfoxide in Relation to Quetiapine Dose, Formulation, and Other Factors. Ther Drug Monit 2012; 34:415-21. [DOI: 10.1097/ftd.0b013e3182603f62] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
30
|
Benyamina A, Naassila M, Bourin M. Potential role of cortical 5-HT(2A) receptors in the anxiolytic action of cyamemazine in benzodiazepine withdrawal. Psychiatry Res 2012; 198:307-12. [PMID: 22421069 DOI: 10.1016/j.psychres.2012.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 12/12/2011] [Accepted: 01/10/2012] [Indexed: 11/19/2022]
Abstract
The antipsychotic cyamemazine is a potent serotonin 5-HT(2A) receptor (5-HT(2AR)) antagonist. A positron emission tomography (PET) study in human patients showed that therapeutic doses of cyamemazine produced near saturation of 5-HT(2AR) occupancy in the frontal cortex, whereas dopamine D(2) occupancy remained below the level for motor side effects observed with typical antipsychotics. Recently, numerous studies have revealed the involvement of 5-HT(2AR) in the pathophysiology of anxiety and a double-blind, randomized clinical trial showed similar efficacy of cyamemazine and bromazepam in reducing the anxiety associated with benzodiazepine withdrawal. Therefore, we reviewed the above articles about 5-HT(2AR) and anxiety in order to understand better the anxiolytic mechanisms of cyamemazine in benzodiazepine withdrawal. The 5-HT(2AR) is the most abundant serotonin receptor subtype in the cortex. Non-pharmacological studies with antisense oligodeoxynucleotides and genetically modified mice clearly showed that cortical 5-HT(2AR) signaling positively modulates anxiety-like behavior. With a few exceptions, most other studies reviewed here further support this view. Therefore, the anxiolytic efficacy of cyamemazine in benzodiazepine withdrawal can be due to a 5-HT(2AR) antagonistic activity at the cortical level.
Collapse
Affiliation(s)
- Amine Benyamina
- Inserm U669, University Hospital Paul Brousse, Villejuif, France.
| | | | | |
Collapse
|
31
|
Abstract
A variety of serotonin (5-HT) receptors, especially 5-HT(2A), 5-HT(1A), 5-HT(6), 5-HT(7), and 5-HT(2C), have been postulated to contribute to the mechanism of action of atypical antipsychotic drugs (APDs), i.e., APDs which cause fewer extrapyramidal side effects (EPS) at clinically optimal doses, in contrast with typical APDs, which are more likely to cause EPS. This advantage, rarely disputed, has made such drugs the preferred treatment for schizophrenia and other indications for APDs. These 5-HT receptors are still of interest as components of novel multireceptor or stand-alone APDs, and potentially to remediate cognitive deficits in schizophrenia. Almost all currently available atypical APDs are 5-HT(2A) receptor inverse agonists, as well as dopamine (DA) D(2) receptor antagonists or partial agonists. Amisulpride, an exceptional atypical APD, has 5-HT(7) antagonism to complement its DA D(2/3) antagonism. Some atypical APDs are also 5-HT(1A) partial agonists, 5-HT(6), or 5-HT(7) antagonists, or some combination of the above. 5-HT(2C) antagonism has been found to contribute to the metabolic side effects of some atypical APDs, whereas 5-HT(2C) agonists have potential as stand-alone APDs and/or cognitive enhancers. This review will provide an update of current preclinical and clinical evidence for the role of these five 5-HT receptors in the actions of current APDs and for the development of novel psychotropic drugs.
Collapse
Affiliation(s)
- Herbert Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
32
|
Ebdrup BH, Rasmussen H, Arnt J, Glenthøj B. Serotonin 2A receptor antagonists for treatment of schizophrenia. Expert Opin Investig Drugs 2011; 20:1211-23. [PMID: 21740279 DOI: 10.1517/13543784.2011.601738] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION All approved antipsychotic drugs share an affinity for the dopamine 2 (D(2)) receptor; however, these drugs only partially ameliorate the symptoms of schizophrenia. It is, therefore, of paramount importance to identify new treatment strategies for schizophrenia. AREAS COVERED Preclinical, clinical and post-mortem studies of the serotonin 5-HT(2A) system in schizophrenia are reviewed. The implications of a combined D(2) and 5-HT(2A) receptor blockade, which is obtained by several current antipsychotic drugs, are discussed, and the rationale for the development of more selective 5-HT(2A) receptor antagonists is evaluated. Moreover, the investigational pipeline of major pharmaceutical companies is examined and an Internet search conducted to identify other pharmaceutical companies investigating 5-HT(2A) receptor antagonists for the treatment of schizophrenia. EXPERT OPINION 5-HT(2A) receptor antagonists appear to assume an intermediate position by being marginally superior to placebo but inferior to conventional antipsychotic drugs. Three previous 5-HT(2A) receptor antagonists have been discontinued after Phase II or III trials, and available Phase IIa data on the remaining 5-HT(2A) receptor antagonist will need substantial additional validation to be approved as a new treatment strategy against schizophrenia.
Collapse
Affiliation(s)
- Bjørn H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research, Copenhagen University Hospital Glostrup, Faculty of Health Sciences, Psychiatric Center Glostrup, Nordre Ringvej 29, DK-2600 Glostrup, Denmark
| | | | | | | |
Collapse
|
33
|
Paterson LM, Kornum BR, Nutt DJ, Pike VW, Knudsen GM. 5-HT radioligands for human brain imaging with PET and SPECT. Med Res Rev 2011; 33:54-111. [PMID: 21674551 DOI: 10.1002/med.20245] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The serotonergic system plays a key modulatory role in the brain and is the target for many drug treatments for brain disorders either through reuptake blockade or via interactions at the 14 subtypes of 5-HT receptors. This review provides the history and current status of radioligands used for positron emission tomography (PET) and single photon emission computerized tomography (SPECT) imaging of human brain serotonin (5-HT) receptors, the 5-HT transporter (SERT), and 5-HT synthesis rate. Currently available radioligands for in vivo brain imaging of the 5-HT system in humans include antagonists for the 5-HT(1A), 5-HT(1B), 5-HT(2A), and 5-HT(4) receptors, and for SERT. Here we describe the evolution of these radioligands, along with the attempts made to develop radioligands for additional serotonergic targets. We describe the properties needed for a radioligand to become successful and the main caveats. The success of a PET or SPECT radioligand can ultimately be assessed by its frequency of use, its utility in humans, and the number of research sites using it relative to its invention date, and so these aspects are also covered. In conclusion, the development of PET and SPECT radioligands to image serotonergic targets is of high interest, and successful evaluation in humans is leading to invaluable insight into normal and abnormal brain function, emphasizing the need for continued development of both SPECT and PET radioligands for human brain imaging.
Collapse
Affiliation(s)
- Louise M Paterson
- Neuropsychopharmacology Unit, Division of Experimental Medicine, Imperial College London, Burlington Danes Building, Du Cane Road, London, United Kingdom
| | | | | | | | | |
Collapse
|
34
|
Meltzer HY, Massey BW. The role of serotonin receptors in the action of atypical antipsychotic drugs. Curr Opin Pharmacol 2011; 11:59-67. [PMID: 21420906 DOI: 10.1016/j.coph.2011.02.007] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 02/22/2011] [Indexed: 12/15/2022]
Abstract
The main class of atypical antipsychotic drugs (APDs) in current use includes the protypical atypical APD, clozapine, as well as aripiprazole, asenapine, iloperidone, lurasidone, olanzapine, quetiapine, risperidone, and ziprasidone. At clinically effective doses, these agents produce extensive blockade of serotonin (5-HT)(2A) receptors, direct or indirect stimulation of 5-HT(1A) receptors, and to a lesser extent, reduction in dopamine (DA) D(2) receptor-mediated neurotransmission. This contrasts with typical APDs, for example haloperidol and perphenazine, which are mainly DA D(2/)D(3) receptor antagonists and have weaker, if any, potency as 5-HT(2A) receptor antagonists. Some, but not all, atypical APDs are also effective 5-HT(2C) receptor inverse agonists or neutral antagonists, 5-HT(6) or 5-HT(7) receptor antagonists. This diverse action on 5-HT receptors may contribute to significant differences in efficacy and tolerability among the atypical APDs. There is considerable preclinical and some clinical evidence that effects on 5-HT receptors contribute to the low risk of producing extrapyramidal side effects, which is the defining characteristic of an atypical APD, the lack of elevation in plasma prolactin levels (with risperidone and 9-hydroxyrisperidone being exceptions), antipsychotic action, and ability to improve some domains of cognition in patients with schizophrenia. The serotonergic actions of the atypical APDs, especially 5-HT(2A) receptor antagonism, are particularly important to the differential effects of typical and atypical APDs to overcome the effects of acute or subchronic administration of N-methyl-d-aspartate (NMDA) receptor antagonists, such as phencyclidine, ketamine, and dizocipline (MK-801). 5-HT(1A) receptor stimulation and 5-HT(6) and 5-HT(7) receptor antagonism may contribute to beneficial effects of these agents on cognition. In particular, 5-HT(7) receptor antagonism may be the basis for the pro-cognitive effects of the atypical APD, amisulpride, a D(2)/D(3) receptor antagonist, which has no effect on other 5-HT receptor. 5-HT(2C) receptor antagonism appears to contribute to the weight gain produced by some atypical APDs and may also affect cognition and psychosis via its influence on cortical and limbic dopaminergic activity.
Collapse
Affiliation(s)
- H Y Meltzer
- Vanderbilt University School of Medicine, Department of Psychiatry, 1601 23rd Avenue South, Suite 306, Nashville, TN 37212, USA.
| | | |
Collapse
|