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
|
Krejčí V, Murínová I, Slanař O, Šíma M. Evidence for Therapeutic Drug Monitoring of Atypical Antipsychotics. Prague Med Rep 2024; 125:101-129. [PMID: 38761044 DOI: 10.14712/23362936.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2024] Open
Abstract
Second-generation antipsychotics (SGAs), also known as atypical antipsychotics, are a newer class of antipsychotic drugs used to treat schizophrenia, bipolar disorder, and related psychiatric conditions. The plasma concentration of antipsychotic drugs is a valid measure of the drug at its primary target structure in the brain, and therefore determines the efficacy and safety of these drugs. However, despite the well-known high variability in pharmacokinetics of these substances, psychiatric medication is usually administered in uniform dosage schedules. Therapeutic drug monitoring (TDM), as the specific method that can help personalised medicine in dose adjustment according to the characteristics of the individual patient, minimizing the risk of toxicity, monitoring adherence, and increasing cost-effectiveness in the treatment, thus seems to be an elegant tool to solve this problem. Non-response to therapeutic doses, uncertain adherence to medication, suboptimal tolerability, or pharmacokinetic drug-drug interactions are typical indications for TDM of SGAs. This review aims to summarize an overview of the current knowledge and evidence of the possibilities to tailor the dosage of selected SGAs using TDM, including the necessary pharmacokinetic parameters for personalised pharmacotherapy.
Collapse
Affiliation(s)
- Veronika Krejčí
- Department of Clinical Pharmacy, Military University Hospital Prague, Prague, Czech Republic.
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - Irena Murínová
- Department of Applied Pharmacy, Faculty of Pharmacy, Masaryk University, Brno, Czech Republic
- Department of Clinical Pharmacy, Military University Hospital Prague, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Martin Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
4
|
Hart XM, Schmitz CN, Gründer G. Molecular Imaging of Dopamine Partial Agonists in Humans: Implications for Clinical Practice. Front Psychiatry 2022; 13:832209. [PMID: 35463532 PMCID: PMC9020768 DOI: 10.3389/fpsyt.2022.832209] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/11/2022] [Indexed: 12/05/2022] Open
Abstract
Positron emission tomography (PET) has been used since the late 1980s for the assessment of relationships between occupancy of D2/3 receptors by antipsychotic drugs in the human brain and the clinical effects and side effects of these compounds in patients. It is now well established for most D2/3 antagonists, both of the first and the second generation, that the ideal occupancy of their target receptors is between approximately 65 and 80%. If the occupancy is below 65%, the probability of treatment response is reduced, if the occupancy is higher than 80%, the risk for extrapyramidal side-effects increases substantially. However, partial agonist antipsychotics behave different from these rules. It has been shown for all three available drugs of this class (aripiprazole, brexpiprazole, cariprazine) that, due to their special pharmacology, a very high target engagement (>90%) not only is not harmful but represents a prerequisite for antipsychotic efficacy. The available PET studies for these drugs are reviewed in this work. It is demonstrated that optimal plasma levels for partial agonist antipsychotics can be derived from these studies, which can guide individual treatment in routine patient care.
Collapse
Affiliation(s)
- Xenia M Hart
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christian N Schmitz
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Gerhard Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
5
|
Eap CB, Gründer G, Baumann P, Ansermot N, Conca A, Corruble E, Crettol S, Dahl ML, de Leon J, Greiner C, Howes O, Kim E, Lanzenberger R, Meyer JH, Moessner R, Mulder H, Müller DJ, Reis M, Riederer P, Ruhe HG, Spigset O, Spina E, Stegman B, Steimer W, Stingl J, Suzen S, Uchida H, Unterecker S, Vandenberghe F, Hiemke C. Tools for optimising pharmacotherapy in psychiatry (therapeutic drug monitoring, molecular brain imaging and pharmacogenetic tests): focus on antidepressants. World J Biol Psychiatry 2021; 22:561-628. [PMID: 33977870 DOI: 10.1080/15622975.2021.1878427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objectives: More than 40 drugs are available to treat affective disorders. Individual selection of the optimal drug and dose is required to attain the highest possible efficacy and acceptable tolerability for every patient.Methods: This review, which includes more than 500 articles selected by 30 experts, combines relevant knowledge on studies investigating the pharmacokinetics, pharmacodynamics and pharmacogenetics of 33 antidepressant drugs and of 4 drugs approved for augmentation in cases of insufficient response to antidepressant monotherapy. Such studies typically measure drug concentrations in blood (i.e. therapeutic drug monitoring) and genotype relevant genetic polymorphisms of enzymes, transporters or receptors involved in drug metabolism or mechanism of action. Imaging studies, primarily positron emission tomography that relates drug concentrations in blood and radioligand binding, are considered to quantify target structure occupancy by the antidepressant drugs in vivo. Results: Evidence is given that in vivo imaging, therapeutic drug monitoring and genotyping and/or phenotyping of drug metabolising enzymes should be an integral part in the development of any new antidepressant drug.Conclusions: To guide antidepressant drug therapy in everyday practice, there are multiple indications such as uncertain adherence, polypharmacy, nonresponse and/or adverse reactions under therapeutically recommended doses, where therapeutic drug monitoring and cytochrome P450 genotyping and/or phenotyping should be applied as valid tools of precision medicine.
Collapse
Affiliation(s)
- C B Eap
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Lausanne, Switzerland, Geneva, Switzerland
| | - G Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - P Baumann
- Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - N Ansermot
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - A Conca
- Department of Psychiatry, Health Service District Bolzano, Bolzano, Italy.,Department of Child and Adolescent Psychiatry, South Tyrolean Regional Health Service, Bolzano, Italy
| | - E Corruble
- INSERM CESP, Team ≪MOODS≫, Service Hospitalo-Universitaire de Psychiatrie, Universite Paris Saclay, Le Kremlin Bicetre, France.,Service Hospitalo-Universitaire de Psychiatrie, Hôpital Bicêtre, Assistance Publique Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - S Crettol
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - M L Dahl
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - J de Leon
- Eastern State Hospital, University of Kentucky Mental Health Research Center, Lexington, KY, USA
| | - C Greiner
- Bundesinstitut für Arzneimittel und Medizinprodukte, Bonn, Germany
| | - O Howes
- King's College London and MRC London Institute of Medical Sciences (LMS)-Imperial College, London, UK
| | - E Kim
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - R Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - J H Meyer
- Campbell Family Mental Health Research Institute, CAMH and Department of Psychiatry, University of Toronto, Toronto, Canada
| | - R Moessner
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - H Mulder
- Department of Clinical Pharmacy, Wilhelmina Hospital Assen, Assen, The Netherlands.,GGZ Drenthe Mental Health Services Drenthe, Assen, The Netherlands.,Department of Pharmacotherapy, Epidemiology and Economics, Department of Pharmacy and Pharmaceutical Sciences, University of Groningen, Groningen, The Netherlands.,Department of Psychiatry, Interdisciplinary Centre for Psychopathology and Emotion Regulation, University of Groningen, Groningen, The Netherlands
| | - D J Müller
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - M Reis
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Clinical Chemistry and Pharmacology, Skåne University Hospital, Lund, Sweden
| | - P Riederer
- Center of Mental Health, Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany.,Department of Psychiatry, University of Southern Denmark Odense, Odense, Denmark
| | - H G Ruhe
- Department of Psychiatry, Radboudumc, Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| | - O 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
| | - E Spina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - B Stegman
- Institut für Pharmazie der Universität Regensburg, Regensburg, Germany
| | - W Steimer
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Munich, Germany
| | - J Stingl
- Institute for Clinical Pharmacology, University Hospital of RWTH Aachen, Germany
| | - S Suzen
- Department of Toxicology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - H Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - S Unterecker
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - F Vandenberghe
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - C Hiemke
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
6
|
Cumming P, Gründer G, Brinson Z, Wong DF. Applications, Advances, and Limitations of Molecular Imaging of Brain Receptors. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
7
|
Grunze H, Csehi R, Born C, Barabássy Á. Reducing Addiction in Bipolar Disorder via Hacking the Dopaminergic System. Front Psychiatry 2021; 12:803208. [PMID: 34970175 PMCID: PMC8712474 DOI: 10.3389/fpsyt.2021.803208] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/23/2021] [Indexed: 02/05/2023] Open
Abstract
The dopaminergic system plays a central and decisive role in substance use disorder (SUD), bipolar disorder (BD), and possibly in a subgroup of patients with refractory depression. Common genetic markers and underlying cellular processes, such as kindling, support the close link between these disorders, which is also expressed by the high rate of comorbidity. Although partial dopamine agonists/antagonists acting on D2 and D3 receptors have an established role in treating BD, their usefulness in SUD is less clear. However, dopamine D3 receptors were shown to play a central role in SUD and BD, making D2/D3 partial agonists/antagonists a potential target for both disorders. This narrative review examines whether these substances bear the promise of a future therapeutic approach especially in patients with comorbid BD and SUD.
Collapse
Affiliation(s)
- Heinz Grunze
- Psychiatrie Schwäbisch Hall, Schwäbisch Hall, Germany
- Paracelsus Medical University, Nuremberg, Germany
| | - Réka Csehi
- Gedeon Richter Plc, Medical Division, Budapest, Hungary
- *Correspondence: Réka Csehi
| | - Christoph Born
- Psychiatrie Schwäbisch Hall, Schwäbisch Hall, Germany
- Paracelsus Medical University, Nuremberg, Germany
| | | |
Collapse
|
8
|
Dopamine D2 Receptor Occupancy Estimated From Plasma Concentrations of Four Different Antipsychotics and the Subjective Experience of Physical and Mental Well-Being in Schizophrenia: Results From the Randomized NeSSy Trial. J Clin Psychopharmacol 2020; 39:550-560. [PMID: 31688449 DOI: 10.1097/jcp.0000000000001131] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Impaired subjective well-being in schizophrenia patients treated with antipsychotics has often been linked inter alia to the antidopaminergic effects of medication. Thus, it is important to capture the association between striatal dopamine D2 receptor occupancy (D2-RO) and global subjective well-being. We examined this association using data from our multicenter, randomized, double-blind Neuroleptic Strategy Study (NeSSy). METHODS An innovative double randomization process was used for allocation of patients to the specific treatment groups. Plasma drug concentrations were measured after 6 and 24 weeks of treatment to obtain the estimated D2-RO (eD2-RO) relative to literature values. We made an exploratory analysis of associations between eD2-RO and subjective well-being scores. One hundred two blood samples from 69 patients were available for the analysis. Because of the lack of a satisfactory occupancy model for quetiapine, only haloperidol, flupentixol, and olanzapine treatment groups were pooled, whereas aripiprazole data were analyzed separately, because of its partial agonistic properties. RESULTS In the pooled antagonist group, eD2-RO correlated negatively with the summarized well-being score. In a more detailed analysis, this association could be confirmed for all first-generation antipsychotic-treated patients, but not for the separate second-generation antipsychotic groups. In the aripiprazole group, higher eD2-RO was associated with impaired physical well-being, but had no association with mental well-being. CONCLUSIONS Our results suggest that high plasma levels and consequently high occupancy at D2 receptors are disadvantageous for subjective well-being, as distinct from the objective extrapyramidal side effects. To minimize patients' malaise, which disfavors adherence, implementation of therapeutic drug monitoring in the clinical routine may be useful.
Collapse
|
9
|
Yan H, Ji JJ, Xiang P, Shen M. Characteristics of quetiapine and 7-hydroxyquetiapine in hair roots and blood after a single dose of quetiapine. Forensic Sci Int 2020; 309:110189. [DOI: 10.1016/j.forsciint.2020.110189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/30/2020] [Accepted: 02/07/2020] [Indexed: 11/25/2022]
|
10
|
Validity and reliability of extrastriatal [11C]raclopride binding quantification in the living human brain. Neuroimage 2019; 202:116143. [DOI: 10.1016/j.neuroimage.2019.116143] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/26/2019] [Accepted: 08/28/2019] [Indexed: 01/25/2023] Open
|
11
|
Affiliation(s)
- Stephen R Marder
- From the Section on Psychosis, Semel Institute for Neuroscience and Human Behavior at the University of California, Los Angeles, and the Veterans Affairs Desert Pacific Mental Illness Research, Education, and Clinical Center, Los Angeles (S.R.M.); and the Departments of Psychology and Psychiatry, Yale University, New Haven, CT (T.D.C.)
| | - Tyrone D Cannon
- From the Section on Psychosis, Semel Institute for Neuroscience and Human Behavior at the University of California, Los Angeles, and the Veterans Affairs Desert Pacific Mental Illness Research, Education, and Clinical Center, Los Angeles (S.R.M.); and the Departments of Psychology and Psychiatry, Yale University, New Haven, CT (T.D.C.)
| |
Collapse
|
12
|
Joo YH, Kim JH, Son YD, Kim HK, Shin YJ, Lee SY, Kim JH. The relationship between excitement symptom severity and extrastriatal dopamine D 2/3 receptor availability in patients with schizophrenia: a high-resolution PET study with [ 18F]fallypride. Eur Arch Psychiatry Clin Neurosci 2018. [PMID: 28623450 DOI: 10.1007/s00406-017-0821-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The purpose of this study was to investigate the relationship between specific symptom severity and D2/3 receptor availability in extrastriatal regions in outpatients with schizophrenia to shed light on the role of extrastriatal dopaminergic neurotransmission in the pathophysiology of symptoms of schizophrenia. Sixteen schizophrenia patients receiving relatively low-dose maintenance atypical antipsychotics and seventeen healthy controls underwent 3-Tesla magnetic resonance imaging and high-resolution positron emission tomography with [18F]fallypride. For D2/3 receptor availability, the binding potential with respect to non-displaceable compartment (BPND) was derived using the simplified reference tissue model. The BPND values were lower in patients on antipsychotic treatment than in controls across all regions with large effect sizes (1.03-1.42). The regions with the largest effect size were the substantia nigra, amygdala, and insula. Symptoms of schizophrenia were assessed using a five-factor model of the Positive and Negative Syndrome Scale (PANSS). The region of interest-based analysis showed that PANSS excitement factor score had a significant positive correlation with the [18F]fallypride BPND in the insula. The equivalent dose of antipsychotics was not significantly correlated with PANSS factor scores or regional BPND values. The voxel-based analysis also revealed a significant positive association between the PANSS excitement factor and the [18F]fallypride BPND in the insula. The present study revealed a significant association between excitement symptom severity and D2/3 receptor availability in the insula in schizophrenia, suggesting a possible important role of D2/3 receptor-mediated neurotransmission in the insula and related limbic system in the pathophysiology of this specific symptom cluster.
Collapse
Affiliation(s)
- Yo-Han Joo
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Jeong-Hee Kim
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Research Institute for Advanced Industrial Technology, Korea University, Sejong, Republic of Korea
| | - Young-Don Son
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon, Republic of Korea
| | - Hang-Keun Kim
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon, Republic of Korea
| | - Yeon-Jeong Shin
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Sang-Yoon Lee
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Department of Radiological Science, College of Health Science, Gachon University, Incheon, Republic of Korea
| | - Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea. .,Department of Psychiatry, Neuroscience Research Institute, Gil Medical Center, Gachon University School of Medicine, Gachon University, 1198 Guwol-dong, Namdong-gu, Incheon, 405-760, Republic of Korea.
| |
Collapse
|
13
|
Kim JH, Cumming P, Son YD, Kim HK, Joo YH, Kim JH. Altered connectivity between striatal and extrastriatal regions in patients with schizophrenia on maintenance antipsychotics: an [18
F]fallypride PET and functional MRI study. Synapse 2018; 72:e22064. [DOI: 10.1002/syn.22064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Jeong-Hee Kim
- Research Institute for Advanced Industrial Technology; Korea University; Sejong Republic of Korea
- Neuroscience Research Institute; Gachon University; Incheon Republic of Korea
| | - Paul Cumming
- School of Psychology and Counselling and IHBI; Queensland University of Technology, and QIMR Berghofer Institute; Brisbane Queensland Australia
| | - Young-Don Son
- Neuroscience Research Institute; Gachon University; Incheon Republic of Korea
- Department of Biomedical Engineering; College of Health Science, Gachon University; Incheon Republic of Korea
| | - Hang-Keun Kim
- Neuroscience Research Institute; Gachon University; Incheon Republic of Korea
- Department of Biomedical Engineering; College of Health Science, Gachon University; Incheon Republic of Korea
| | - Yo-Han Joo
- Neuroscience Research Institute; Gachon University; Incheon Republic of Korea
| | - Jong-Hoon Kim
- Neuroscience Research Institute; Gachon University; Incheon Republic of Korea
- Department of Psychiatry, Gil Medical Center; Gachon University College of Medicine, Gachon University; Incheon Republic of Korea
| |
Collapse
|
14
|
Veselinović T, Vernaleken I, Janouschek H, Cumming P, Paulzen M, Mottaghy FM, Gründer G. The role of striatal dopamine D 2/3 receptors in cognitive performance in drug-free patients with schizophrenia. Psychopharmacology (Berl) 2018; 235:2221-2232. [PMID: 29717334 DOI: 10.1007/s00213-018-4916-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 04/18/2018] [Indexed: 10/17/2022]
Abstract
OBJECTIVE A considerable body of research links cognitive function to dopaminergic transmission in the prefrontal cortex, but less is known about cognition in relation to striatal dopamine D2/3 receptors in unmedicated patients with psychosis. METHODS We investigated this association by obtaining PET recordings with the high-affinity D2/3 antagonist ligand [18F] fallypride in 15 medication-free patients with schizophrenia and 11 healthy controls. On the day of PET scanning, we undertook comprehensive neuropsychological testing and assessment of psychopathology using the Positive and Negative Syndrome Scale (PANSS). RESULTS The patients' performance in cognitive tests was significantly impaired in almost all domains. Irrespective of medication history, the mean [18F] fallypride binding potential (BPND) in the patient group tended to be globally 5-10% higher than that of the control group, but without reaching significance in any brain region. There were significant positive correlations between individual patient performance in the Trail Making Test (TMT(A) and TMT(B)) and Digit-Symbol-Substitution-Test with regional [18F] fallypride BPND, which remained significant after Bonferroni correction for the TMT(A) in caudate nucleus (CN) and for the TMT(B) in CN and putamen. No such correlations were evident in the control group. DISCUSSION The association between better cognitive performance and greater BPND in schizophrenia patients may imply that relatively lower receptor occupancy by endogenous dopamine favors better sparing of cognitive function. Absence of comparable correlations in healthy controls could indicate a greater involvement of signaling at dopamine D2/3 receptors in certain cognitive functions in schizophrenia patients than in healthy controls.
Collapse
Affiliation(s)
- Tanja Veselinović
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany. .,Jülich Aachen Research Alliance JARA, Translational Brain Medicine, Jülich, Germany.
| | - Ingo Vernaleken
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.,Jülich Aachen Research Alliance JARA, Translational Brain Medicine, Jülich, Germany
| | - Hildegard Janouschek
- Department of Psychiatry and Iowa Neuroscience Institute, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Paul Cumming
- School of Psychology and Counselling and IHBI, Queensland University of Technology, and QIMR-Berghofer Institute, Brisbane, Australia
| | - Michael Paulzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.,Jülich Aachen Research Alliance JARA, Translational Brain Medicine, Jülich, Germany.,Alexianer Hospital Aachen, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany.,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Gerhard Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
15
|
Caravaggio F, Fervaha G, Iwata Y, Plitman E, Chung JK, Nakajima S, Mar W, Gerretsen P, Kim J, Chakravarty MM, Mulsant B, Pollock B, Mamo D, Remington G, Graff-Guerrero A. Amotivation is associated with smaller ventral striatum volumes in older patients with schizophrenia. Int J Geriatr Psychiatry 2018; 33:523-530. [PMID: 29110353 PMCID: PMC5807115 DOI: 10.1002/gps.4818] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/08/2017] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Motivational deficits are prevalent in patients with schizophrenia, persist despite antipsychotic treatment, and predict long-term outcomes. Evidence suggests that patients with greater amotivation have smaller ventral striatum (VS) volumes. We wished to replicate this finding in a sample of older, chronically medicated patients with schizophrenia. Using structural imaging and positron emission tomography, we examined whether amotivation uniquely predicted VS volumes beyond the effects of striatal dopamine D2/3 receptor (D2/3 R) blockade by antipsychotics. METHODS Data from 41 older schizophrenia patients (mean age: 60.2 ± 6.7; 11 female) were reanalysed from previously published imaging data. We constructed multivariate linear stepwise regression models with VS volumes as the dependent variable and various sociodemographic and clinical variables as the initial predictors: age, gender, total brain volume, and antipsychotic striatal D2/3 R occupancy. Amotivation was included as a subsequent step to determine any unique relationships with VS volumes beyond the contribution of the covariates. In a reduced sample (n = 36), general cognition was also included as a covariate. RESULTS Amotivation uniquely explained 8% and 6% of the variance in right and left VS volumes, respectively (right: β = -.38, t = -2.48, P = .01; left: β = -.31, t = -2.17, P = .03). Considering cognition, amotivation levels uniquely explained 9% of the variance in right VS volumes (β = -.43, t = -0.26, P = .03). CONCLUSION We replicate and extend the finding of reduced VS volumes with greater amotivation. We demonstrate this relationship uniquely beyond the potential contributions of striatal D2/3 R blockade by antipsychotics. Elucidating the structural correlates of amotivation in schizophrenia may help develop treatments for this presently irremediable deficit.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Gagan Fervaha
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Yusuke Iwata
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Eric Plitman
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Jun Ku Chung
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Shinichiro Nakajima
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Wanna Mar
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Julia Kim
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - M. Mallar Chakravarty
- Department of Biological & Biomedical Engineering, McGill University, Montreal, Quebec, Canada. H4H 1R3
- Cerebral Imaging Centre, Douglas Mental Health Institute, McGill University, Montreal, Quebec, Canada. H4H 1R3
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada. H4H 1R3
| | - Benoit Mulsant
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Bruce Pollock
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - David Mamo
- Department of Psychiatry, University of Malta, Malta
| | - Gary Remington
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, Canada. M5T 1R8
| |
Collapse
|
16
|
Deutschländer A, la Fougère C, Boetzel K, Albert NL, Gildehaus FJ, Bartenstein P, Xiong G, Cumming P. Occupancy of pramipexole (Sifrol) at cerebral dopamine D2/3 receptors in Parkinson's disease patients. NEUROIMAGE-CLINICAL 2016; 12:41-6. [PMID: 27408789 PMCID: PMC4925448 DOI: 10.1016/j.nicl.2016.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 11/19/2022]
Abstract
Whereas positron emission tomography (PET) with the antagonist ligand [18F]fallypride reveals the composite of dopamine D2 and D3 receptors in brain, treatment of Parkinson's disease (PD) patients with the D3-prefering agonist pramipexole should result in preferential occupancy in the nucleus accumbens, where the D3-subtype is most abundant. To test this prediction we obtained pairs of [18F]fallypride PET recordings in a group of nine PD patients, first in a condition of treatment as usual with pramipexole (ON-Sifrol; 3 × 0.7 mg p.d.), and again at a later date, after withholding pramipexole 48–72 h (OFF-Sifrol); in that condition the serum pramipexole concentration had declined by 90% and prolactin levels had increased four-fold, in conjunction with a small but significant worsening of PD motor symptoms. Exploratory comparison with historical control material showed 14% higher dopamine D2/3 availability in the more-affected putamen of patients OFF medication. On-Sifrol there was significant (p ˂ 0.01) occupancy at [18F]fallypride binding sites in globus pallidus (8%) thalamus (9%) and substantia nigra (19%), as well as marginally significant occupancy in frontal and temporal cortex of patients. Contrary to expectation, comparison of ON- and OFF-Sifrol results did not reveal any discernible occupancy in nucleus accumbens, or elsewhere in the extended striatum; present methods should be sensitive to a 10% change in dopamine D2/3 receptor availability in striatum; the significant findings elsewhere in the basal ganglia and in cerebral cortex are consistent with a predominance of D3 receptors in those structures, especially in substantia nigra, and imply that therapeutic effects of pramipexole may be obtained at sites outside the extended striatum. Fallypride PET recordings in nine PD patients, scanned on- and off medication with pramipexole No occupancy in the striatum, despite improved motor symptoms Substantial occupancy in substantia nigra, thalamus and globus pallidus
Collapse
Affiliation(s)
| | | | - Kai Boetzel
- Department of Neurology, Ludwig-Maximilians University of Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilians University of Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians University of Munich, Germany
| | - Guoming Xiong
- Department of Nuclear Medicine, Ludwig-Maximilians University of Munich, Germany
| | - Paul Cumming
- Department of Neuropsychiatry and Psychosomatic Medicine, Rikshospitalet, University of Oslo, Oslo, Norway; School of Psychology and Counselling, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
17
|
Davis RE, Vanover KE, Zhou Y, Brašić JR, Guevara M, Bisuna B, Ye W, Raymont V, Willis W, Kumar A, Gapasin L, Goldwater DR, Mates S, Wong DF. ITI-007 demonstrates brain occupancy at serotonin 5-HT₂A and dopamine D₂ receptors and serotonin transporters using positron emission tomography in healthy volunteers. Psychopharmacology (Berl) 2015; 232:2863-72. [PMID: 25843749 DOI: 10.1007/s00213-015-3922-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 03/17/2015] [Indexed: 01/01/2023]
Abstract
RATIONALE Central modulation of serotonin and dopamine underlies efficacy for a variety of psychiatric therapeutics. ITI-007 is an investigational new drug in development for treatment of schizophrenia, mood disorders, and other neuropsychiatric disorders. OBJECTIVES The purpose of this study was to determine brain occupancy of ITI-007 at serotonin 5-HT2A receptors, dopamine D2 receptors, and serotonin transporters using positron emission tomography (PET) in 16 healthy volunteers. METHODS Carbon-11-MDL100907, carbon-11-raclopride, and carbon-11-3-amino-4-(2-dimethylaminomethyl-phenylsulfanyl)-benzonitrile) (carbon-11-DASB) were used as the radiotracers for imaging 5-HT2A receptors, D2 receptors, and serotonin transporters, respectively. Brain regions of interest were outlined using magnetic resonance tomography (MRT) with cerebellum as the reference region. Binding potentials were estimated by fitting a simplified reference tissue model to the measured tissue-time activity curves. Target occupancy was expressed as percent change in the binding potentials before and after ITI-007 administration. RESULTS Oral ITI-007 (10-40 mg) was safe and well tolerated. ITI-007 rapidly entered the brain with long-lasting and dose-related occupancy. ITI-007 (10 mg) demonstrated high occupancy (>80 %) of cortical 5-HT2A receptors and low occupancy of striatal D2 receptors (~12 %). D2 receptor occupancy increased with dose and significantly correlated with plasma concentrations (r (2) = 0.68, p = 0.002). ITI-007 (40 mg) resulted in peak occupancy up to 39 % of striatal D2 receptors and 33 % of striatal serotonin transporters. CONCLUSIONS The results provide evidence for a central mechanism of action via dopaminergic and serotonergic pathways for ITI-007 in living human brain and valuable information to aid dose selection for future clinical trials.
Collapse
Affiliation(s)
- Robert E Davis
- Intra-Cellular Therapies, Inc. (ITI), 3960 Broadway, 6th floor, New York, NY, 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Skov L, Johansen SS, Linnet K. Postmortem Quetiapine Reference Concentrations in Brain and Blood. J Anal Toxicol 2015; 39:557-61. [DOI: 10.1093/jat/bkv072] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
19
|
Rominger A, Cumming P, Brendel M, Xiong G, Zach C, Karch S, Tatsch K, Bartenstein P, la Fougère C, Koch W, Pogarell O. Altered serotonin and dopamine transporter availabilities in brain of depressed patients upon treatment with escitalopram: A [123 I]β-CIT SPECT study. Eur Neuropsychopharmacol 2015; 25:873-81. [PMID: 25819144 DOI: 10.1016/j.euroneuro.2014.12.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 11/25/2014] [Accepted: 12/24/2014] [Indexed: 01/13/2023]
Abstract
Altered SERT and DAT availabilities during treatment with escitalopram were investigated with [(123)I]2β-carbomethoxy-3β-(4-iodophenyl)tropane (β-CIT) SPECT in a series of patients fulfilling the criteria for unipolar major depressive disorder (MDD). 27 patients (10m, 42±16y) with diagnosis of MDD were recruited for the study. All patients underwent neuropsychiatric testing for assessment of Hamilton Depression (HAM-D) and Beck Depression Inventory (BDI) scores. At baseline, [(123)I]β-CIT SPECT recordings were acquired 4h (SERT-weighted) and 20-24h p.i (DAT-weighted). Follow-up scans and neuropsychiatric testing were performed after six weeks of stable escitalopram medication. Voxel-wise parametric maps of specific/ non-specific ratios-1 (~BPND) were calculated. At baseline, DAT-weighted BPND was 5.06±0.81 in striatum and SERT-weighted BPND was 0.94±0.18 in thalamus. There were significant negative correlations with age for DAT in striatum (R=-0.60; p<0.01) and SERT in thalamus (R=-0.45; p<0.05). Under SSRI treatment there was an apparent 42% occupancy of SERT in thalamus (p<0.0001), whereas DAT availability increased significantly by 20% in striatum (p<0.001); higher apparent SERT occupancy in thalamus was associated with lesser DAT increase in striatum (R=-0.62; p<0.005). The low apparent SERT occupancy may be confounded by alterations in SERT expression during treatment. Thus, [(123)I]β-CIT SPECT revealed age-dependent declines in DAT and SERT availabilities in un-medicated MDD patients, comparable to that seen previously in healthy controls. At follow-up, the SSRI-evoked increase in DAT was less pronounced in the older patients, even though apparent SERT occupancy and clinical improvement were not age-dependent. Present findings may have implications for escitalopram dosage and side effect profile in younger MDD patients.
Collapse
Affiliation(s)
- A Rominger
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany.
| | - P Cumming
- Department of Psychiatry, University of Oslo, Oslo, Norway
| | - M Brendel
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - G Xiong
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - C Zach
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - S Karch
- Department of Psychiatry, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - K Tatsch
- Department of Nuclear Medicine, Municipal Hospital Karlsruhe Inc., Karlsruhe, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - C la Fougère
- Department of Nuclear Medicine, University of Tubingen, Tubingen, Germany
| | - W Koch
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - O Pogarell
- Department of Psychiatry, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
20
|
|
21
|
Piel M, Vernaleken I, Rösch F. Positron Emission Tomography in CNS Drug Discovery and Drug Monitoring. J Med Chem 2014; 57:9232-58. [DOI: 10.1021/jm5001858] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Markus Piel
- Institute
of Nuclear Chemistry, Johannes Gutenberg-University, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| | - Ingo Vernaleken
- Department
of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University, Pauwelsstraße 30, D-52074 Aachen, Germany
| | - Frank Rösch
- Institute
of Nuclear Chemistry, Johannes Gutenberg-University, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| |
Collapse
|
22
|
Dunn JT, Clark-Papasavas C, Marsden P, Baker S, Cleij M, Kapur S, Kessler R, Howard R, Reeves SJ. Establishing test-retest reliability of an adapted [(18)F]fallypride imaging protocol in older people. J Cereb Blood Flow Metab 2013; 33:1098-103. [PMID: 23591647 PMCID: PMC3705439 DOI: 10.1038/jcbfm.2013.55] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/21/2013] [Accepted: 03/15/2013] [Indexed: 02/04/2023]
Abstract
[(18)F]fallypride is a high-affinity dopamine D2/3 receptor tracer with the ability to reliably quantify D2/3 receptor sites in both striatal and corticolimbic regions. The translational potential of [(18)F]fallypride imaging is, however, limited by the lengthy scanning sessions (60-80 minutes duration over a total of 3-4 hours) required by current protocols. The aims of our study were to adapt [(18)F]fallypride imaging for use in clinical populations with neurological and neuropsychiatric disorders, by reducing the duration of individual scanning sessions; and to establish the reproducibility and reliability of our adapted protocol in healthy older people. Eight participants (five male and three female; mean age=75.87±4.39 years) were scanned twice, 4-6 weeks apart. [(18)F]fallypride binding potential was determined from image data collected during three sampling times: 0-30; 60-90; and 210-240 minutes post injection. High reproducibility and reliability (test-retest variability <8%; intraclass correlation coefficient >0.8) were observed in all but the prefrontal regions, and remained so when sampling times were reduced to 20 minutes (0-20; 70-90; 220-240 minutes). The adapted protocol is feasible for use across neuropsychiatric disorders in which dopamine has been implicated and is sufficiently sensitive to detect within-subject changes between 2.7% and 5.5% in striatal and limbic regions.
Collapse
Affiliation(s)
- Joel T Dunn
- St Thomas' PET Imaging Centre, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Vulnerability to psychotogenic effects of ketamine is associated with elevated D2/3-receptor availability. Int J Neuropsychopharmacol 2013; 16:745-54. [PMID: 22906553 DOI: 10.1017/s1461145712000764] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Previous positron emission tomography (PET) studies employing competition paradigms have shown either no change or substantial declines in striatal [(11)C]-raclopride binding after challenge with psychotogenic doses of the N-methyl-D-aspartate antagonist ketamine. We sought to probe the relationship between the severity of ketamine-induced psychotic symptoms and altered dopamine D(2/3) receptor availability throughout brain using the high affinity ligand [(18)F]-fallypride (FP). PET recordings were obtained in a group of 10 healthy, young male volunteers, in a placebo condition, and in the course of an infusion with ketamine at a psychotomimetic dose. Administration of the Positive and Negative Syndrome Scale and the Thought and Language Index in both conditions revealed a substantial emergence of mainly negative symptoms of schizophrenia, persisting until the end of the 3 h PET recordings. The baseline FP binding in cortex, caudate nucleus and other brain regions was highly predictive of the individual severity of psychotic symptoms in the ketamine condition. However, there was no evidence of ketamine-evoked reductions in FP binding. In the context of earlier findings, we speculate that high baseline D(2/3)-receptor availability may impart benefits with regard to cognitive flexibility, but increases the risk of maladaptive information processing in the face of environmental stresses and challenges.
Collapse
|
24
|
Cumming P, Xiong G, la Fougère C, Rominger A, Bartenstein P, Buchholz HG, Piel M, Rösch F, Gründer G, Vernaleken I. Surrogate markers for cerebral blood flow correlate with [¹⁸F]-fallypride binding potential at dopamine D(2/3) receptors in human striatum. Synapse 2013; 67:199-203. [PMID: 23239525 DOI: 10.1002/syn.21630] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 12/05/2012] [Accepted: 12/07/2012] [Indexed: 12/29/2022]
Abstract
Positron emission tomography (PET) with the high affinity dopamine D(2/3) receptor ligand [¹⁸F]-fallypride affords estimates of the binding potential (BP(ND) ) in extra-striatal regions of low receptor abundance, but the sufficient recording time for accurate measurements in striatum has been called into question. We have earlier argued that transient equilibrium measurements are obtained in striatum with [¹⁸F]-fallypride PET recordings of 3 h duration, which may be the practical limit for clinical investigations without interrupted scanning. However, the high extraction fraction of [¹⁸F]-fallypride predicts flow-dependence of tracer delivery to brain, which may be a source of variance of the apparent BP(ND) in regions of high binding. To test this prediction, we conducted a retrospective analysis of [¹⁸F]-fallypride PET data from a group of 50 healthy volunteers (age 18-58 years [mean ± SD: 32.6 ± 10.6), who had participated in clinical studies without arterial input measurements. We used the initial 120-s integral (AUC) of the venous confluence (VC) as a surrogate marker for cerebral blood flow (CBF) and tested for correlations between regional estimates of BP(ND) calculated by the simplified reference tissue model (SRTM) and the individual VC-AUC. The magnitude of BP(ND) in a high binding region (putamen), but not in a low binding region (thalamus) correlated positively with VC-AUC, suggesting that approximately 9% of the variance in the [¹⁸F]-fallypride BP(ND) in putamen can be attributed to individual differences in this surrogate marker for CBF, a contribution equal in magnitude to the effects of age on BP(ND) in putamen of the present healthy control group.
Collapse
Affiliation(s)
- Paul Cumming
- Department of Nuclear Medicine, Ludwig-Maximilian University, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
te Beek ET, de Boer P, Moerland M, Schmidt ME, Hoetjes NJ, Windhorst AD, van Berckel BNM, Cohen AF, van Gerven JMA, Lammertsma AA. In vivo quantification of striatal dopamine D2 receptor occupancy by JNJ-37822681 using [11C]raclopride and positron emission tomography. J Psychopharmacol 2012; 26:1128-35. [PMID: 22290934 DOI: 10.1177/0269881111435251] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
JNJ-37822681 is a novel, fast-dissociating dopamine D(2) receptor antagonist, currently in development as an antipsychotic drug candidate. A previous first-in-human study demonstrated mild central nervous system effects of JNJ-37822681 in healthy male volunteers. Significant but transient serum prolactin elevations were demonstrated, whereas other neurophysiological effects were relatively small. To investigate striatal dopamine D(2) receptor occupancy by variable single doses of JNJ-37822681, an open-label [(11)C]raclopride positron emission tomography study was performed in 12 healthy male volunteers, using the simplified reference tissue model with cerebellum as reference tissue. Oral administration of JNJ-37822681 resulted in dose-dependent dopamine D(2) receptor occupancy. Receptor occupancy increased from 9-19% at 2 mg doses to 60-74% at 20 mg doses of JNJ-37822681. Therefore, single oral doses of JNJ-37822681 can produce occupancy levels that are generally associated with clinical efficacy for registered antipsychotic drugs.
Collapse
Affiliation(s)
- Erik T te Beek
- Centre for Human Drug Research, Zernikedreef 10, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
In vivo evidence of deep brain stimulation-induced dopaminergic modulation in Tourette's syndrome. Biol Psychiatry 2012; 71:e11-3. [PMID: 22129758 DOI: 10.1016/j.biopsych.2011.09.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 09/30/2011] [Accepted: 09/30/2011] [Indexed: 12/15/2022]
|
27
|
Abstract
This review summarizes the current state of knowledge regarding the proposed mechanisms by which antipsychotic agents reduce the symptoms of schizophrenia while giving rise to adverse side effects. The first part summarizes the contribution of neuroimaging studies to our understanding of the neurochemical substrates of schizophrenia, putting emphasis on direct evidence suggestive of a presynaptic rather than a postsynaptic dysregulation of dopaminergic neurotransmission in this disorder. The second part addresses the role of D(2) and non-D(2) receptor blockade in the treatment of schizophrenia and highlights a preponderant role of D(2) receptors in the mechanism of antipsychotic action. Neuroimaging studies have defined a narrow, but optimal, therapeutic window of 65-78 % D(2) receptor blockade within which most antipsychotics achieve optimal clinical efficacy with minimal side effects. Some antipsychotics though do not conform to that therapeutic window, notably clozapine. The reasons for its unexcelled clinical efficacy despite subthreshold levels of D(2) blockade are unclear and current theories on clozapine's mechanisms of action are discussed, including transiency of its D(2) receptor blocking effects or preferential blockade of limbic D(2) receptors. Evidence is also highlighted to consider the use of extended antipsychotic dosing to achieve transiency of D(2) blockade as a way to optimize functional outcomes in patients. We also present some critical clinical considerations regarding the mechanisms linking dopamine disturbance to the expression of psychosis and its blockade to the progressive resolution of psychosis, keeping in perspective the speed and onset of antipsychotic action. Finally, we discuss potential novel therapeutic strategies for schizophrenia.
Collapse
Affiliation(s)
- Nathalie Ginovart
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| | | |
Collapse
|
28
|
Abstract
The high-affinity radioligand [(18)F]fallypride (FP) is frequently used for quantification of striatal/extrastriatal D(2/3) receptors and the receptor occupancies of antipsychotics (APs). Its 110 minutes half-life allows long scan durations. However, the optimum scan duration is a matter of debate. This investigation focuses on scan-duration-related effects on simplified reference tissue model (SRTM) results and the time point of transient equilibrium in a large sample of dynamic FP positron emission tomography (PET) scans. Fifty drug-free and 50 AP-treated subjects underwent FP-PET scans (180 minutes scan duration). The binding potential (BP(ND)) of the putamen, thalamus, and temporal cortex were calculated using the SRTM and the transient equilibrium model. Furthermore, receptor occupancies were calculated for AP-treated patients. Transient equilibrium in the unblocked putamen occurred after 121±29.6 minutes. The transient equilibrium occurred much earlier in the extrastriatal regions or under AP treatment. Stepwise scan shortening caused BP(ND) underestimations of 0.58% for the first 10-minute reduction (putamen, SRTM), finally reaching 5.76% after 1 hour scan-time reduction. We observed preferential extrastriatal AP binding irrespective of the analytical method. [(18)F]fallypride scan durations of 180 minutes reliably reach equilibrium even in D(2/3)-receptor-rich regions. Moderate reductions in FP scan durations only caused small changes to SRTM results even in receptor-rich regions. Apparently, the D(2/3) receptor occupancy results of APs, especially preferential extrastriatal binding observations, are not relevantly biased by inappropriate scan durations.
Collapse
|