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
|
Howes OD, Dawkins E, Lobo MC, Kaar SJ, Beck K. New Drug Treatments for Schizophrenia: A Review of Approaches to Target Circuit Dysfunction. Biol Psychiatry 2024; 96:638-650. [PMID: 38815885 DOI: 10.1016/j.biopsych.2024.05.014] [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: 12/29/2023] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024]
Abstract
Schizophrenia is a leading cause of global disease burden. Current drug treatments are associated with significant side effects and have limited efficacy for many patients, highlighting the need to develop new approaches that target other aspects of the neurobiology of schizophrenia. Preclinical, in vivo imaging, postmortem, genetic, and pharmacological studies have highlighted the key role of cortical GABAergic (gamma-aminobutyric acidergic)-glutamatergic microcircuits and their projections to subcortical dopaminergic circuits in the pathoetiology of negative, cognitive, and psychotic symptoms. Antipsychotics primarily act downstream of the dopaminergic component of this circuit. However, multiple drugs are currently in development that could target other elements of this circuit to treat schizophrenia. These include drugs for GABAergic or glutamatergic targets, including glycine transporters, D-amino acid oxidase, sodium channels, or potassium channels. Other drugs in development are likely to primarily act on pathways that regulate the dopaminergic system, such as muscarinic or trace amine receptors or 5-HT2A receptors, while PDE10A inhibitors are being developed to modulate the downstream consequences of dopaminergic dysfunction. Our review considers where new drugs may act on this circuit and their latest clinical trial evidence in terms of indication, efficacy, and side effects. Limitations of the circuit model, including whether there are neurobiologically distinct subgroups of patients, and future directions are also considered. Several drugs based on the mechanisms reviewed have promising clinical data, with the muscarinic agonist KarXT most advanced. If these drugs are approved for clinical use, they have the potential to revolutionize understanding of the pathophysiology and treatment of schizophrenia.
Collapse
Affiliation(s)
- Oliver D Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom.
| | - Eleanor Dawkins
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom
| | - Maria C Lobo
- South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom
| | - Stephen J Kaar
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Division of Psychology and Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Greater Manchester Mental Health National Health Service Foundation Trust, Manchester, United Kingdom
| | - Katherine Beck
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom
| |
Collapse
|
3
|
Liu L, Li Z, Kong D, Huang Y, Wu D, Zhao H, Gao X, Zhang X, Yang M. Neuroimaging markers of aberrant brain activity and treatment response in schizophrenia patients based on brain complexity. Transl Psychiatry 2024; 14:365. [PMID: 39251595 PMCID: PMC11384759 DOI: 10.1038/s41398-024-03067-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024] Open
Abstract
The complexity of brain activity reflects its ability to process information, adapt to environmental changes, and transition between states. However, it remains unclear how schizophrenia (SZ) affects brain activity complexity, particularly its dynamic changes. This study aimed to investigate the abnormal patterns of brain activity complexity in SZ, their relationship with cognitive deficits, and the impact of antipsychotic medication. Forty-four drug-naive first-episode (DNFE) SZ patients and thirty demographically matched healthy controls (HC) were included. Functional MRI-based sliding window analysis was utilized for the first time to calculate weighted permutation entropy to characterize complex patterns of brain activity in SZ patients before and after 12 weeks of risperidone treatment. Results revealed reduced complexity in the caudate, putamen, and pallidum at baseline in SZ patients compared to HC, with reduced complexity in the left caudate positively correlated with Continuous Performance Test (CPT) and Category Fluency Test scores. After treatment, the complexity of the left caudate increased. Regions with abnormal complexity showed decreased functional connectivity, with complexity positively correlated with connectivity strength. We observed that the dynamic complexity of the brain exhibited the characteristic of spontaneous, recurring "complexity drop", potentially reflecting transient state transitions in the resting brain. Compared to HC, patients exhibited reduced scope, intensity, and duration of complexity drop, all of which improved after treatment. Reduced duration was negatively correlated with CPT scores and positively with clinical symptoms. The results suggest that abnormalities in brain activity complexity and its dynamic changes may underlie cognitive deficits and clinical symptoms in SZ patients. Antipsychotic treatment partially restores these abnormalities, highlighting their potential as indicators of treatment efficacy and biomarkers for personalized therapy.
Collapse
Affiliation(s)
- Liju Liu
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Zezhi Li
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Di Kong
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yanqing Huang
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Diwei Wu
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Huachang Zhao
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xin Gao
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiangyang Zhang
- Affiliated Mental Health Center of Anhui Medical University; Hefei Fourth People's Hospital; Anhui Mental Health Center, Hefei, PR China.
| | - Mi Yang
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China.
| |
Collapse
|
4
|
O'Neill JR, Jameson A, McLean SL, Dixon M, Cardno AG, Lawrence C. A proposal for reducing maximum target doses of drugs for psychosis: Reviewing dose-response literature. J Psychopharmacol 2024; 38:344-352. [PMID: 38494791 DOI: 10.1177/02698811241239543] [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] [Indexed: 03/19/2024]
Abstract
BACKGROUND Presently, there is limited guidance on the maximal dosing of psychosis drugs that is based on effectiveness rather than safety or toxicity. Current maximum dosing recommendations may far exceed the necessary degree of dopamine D2 receptor blockade required to treat psychosis. This may lead to excess harm through cognitive impairment and side effects. AIMS This analysis aimed to establish guidance for prescribers by optimally dosing drugs for psychosis based on efficacy and benefit. METHODS We used data from two dose-response meta-analyses and reviewed seven of the most prescribed drugs for psychosis in the UK. Where data were not available, we used appropriate comparison techniques based on D2 receptor occupancy to extrapolate our recommendations. RESULTS We found that the likely threshold dose for achieving remission of psychotic symptoms was often significantly below the currently licensed dose for these drugs. We therefore recommend that clinicians are cautious about exceeding our recommended doses. Individual factors, however, should be accounted for. We outline potentially relevant factors including age, ethnicity, sex, smoking status and pharmacogenetics. Additionally, we recommend therapeutic drug monitoring as a tool to determine individual pharmacokinetic variation. CONCLUSIONS In summary, we propose a new set of maximum target doses for psychosis drugs based on efficacy. Further research through randomised controlled trials should be undertaken to evaluate the effect of reducing doses from current licensing maximums or from doses that are above our recommendations. However, dose reductions should be implemented in a manner that accounts for and reduces the effects of drug withdrawal.
Collapse
Affiliation(s)
- James R O'Neill
- Faculty of Medicine and Health, University of Leeds, Leeds, UK
- South West Yorkshire Partnership NHS Foundation Trust, Wakefield, UK
- Leeds and York Partnership NHS Foundation Trust, Leeds, UK
| | - Adam Jameson
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
- Wolfson Centre for Applied Health Research, Bradford, UK
- Bradford District Care NHS Foundation Trust, Shipley, UK
| | - Samantha L McLean
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
- Wolfson Centre for Applied Health Research, Bradford, UK
| | - Michael Dixon
- Leeds and York Partnership NHS Foundation Trust, Leeds, UK
| | | | - Christopher Lawrence
- Southern Health NHS Foundation Trust, Southampton, UK
- University of Southampton, Southampton, UK
| |
Collapse
|
5
|
Bandín-Vilar E, Toja-Camba FJ, Vidal-Millares M, Durán-Maseda MJ, Pou-Álvarez M, Castro-Balado A, Maroñas O, Gil-Rodríguez A, Carracedo Á, Zarra-Ferro I, Soy D, Fernández-Ferreiro A, Mangas-Sanjuan V, Mondelo-García C. Towards precision medicine of long-acting aripiprazole through population pharmacokinetic modelling. Psychiatry Res 2024; 333:115721. [PMID: 38245977 DOI: 10.1016/j.psychres.2024.115721] [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: 10/11/2023] [Revised: 12/02/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024]
Abstract
Population pharmacokinetic (popPK) models constitute a valuable tool for characterizing the pharmacokinetic properties of once-monthly long-acting injectable aripiprazole (LAI aripiprazole) and quantifying the sources of variability in drug exposure. Our aim is to develop a popPK model of both aripiprazole and its metabolite dehydro-aripiprazole in patients treated with LAI aripiprazole, and to personalize the dosing regimen of aripiprazole across different sub-groups of patients. This is a prospective study investigating the pharmacokinetics of LAI aripiprazole. A total of 93 patients were included, 21 for model development and 71 for external model evaluation. A one-compartment model with linear absorption and elimination adequately described both aripiprazole and dehydro-aripiprazole concentrations. The weight of the patients has been shown to be the factor that most influences the absorption. However, the metabolizing phenotype for CYP2D6 and the concomitant treatment with strong inhibitors of this cytochrome have been shown to be the covariates that most influence total drug exposure. This is the first popPK model developed for LAI aripiprazole that includes aripiprazole and its main active metabolite, dehydroaripiprazole. It provides a personalized dosage recommendation that maximizes the probability of achieving optimal therapeutic concentrations and minimizes the difficulties associated with trial-and-error therapeutic strategies carried out in clinical practice.
Collapse
Affiliation(s)
- Enrique Bandín-Vilar
- Pharmacy Department, University Clinical Hospital Santiago de Compostela (CHUS), Spain; Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Spain; Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, University of Santiago de Compostela (USC), Spain
| | - Francisco José Toja-Camba
- Pharmacy Department, University Clinical Hospital Santiago de Compostela (CHUS), Spain; Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Spain; Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, University of Santiago de Compostela (USC), Spain
| | - María Vidal-Millares
- Psychiatry Department, University Clinical Hospital of Santiago de Compostela, Spain
| | | | - Marta Pou-Álvarez
- Psychiatry Department, University Clinical Hospital of Santiago de Compostela, Spain
| | - Ana Castro-Balado
- Pharmacy Department, University Clinical Hospital Santiago de Compostela (CHUS), Spain; Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Spain; Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, University of Santiago de Compostela (USC), Spain
| | - Olalla Maroñas
- Genomic Medicine Group CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; Galician Foundation of Genomic Medicine, Foundation of Health Research Institute of Santiago de Compostela (FIDIS), SERGAS, Santiago de Compostela, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain; Pharmacogenomics and drug discovery, Health Research Institute of Santiago de Compostela (IDIS), Spain
| | - Almudena Gil-Rodríguez
- Genomic Medicine Group CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; Pharmacogenomics and drug discovery, Health Research Institute of Santiago de Compostela (IDIS), Spain
| | - Ángel Carracedo
- Galician Foundation of Genomic Medicine, Foundation of Health Research Institute of Santiago de Compostela (FIDIS), SERGAS, Santiago de Compostela, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, Madrid, Spain; Pharmacogenomics and drug discovery, Health Research Institute of Santiago de Compostela (IDIS), Spain; Genetics group, Health Research Institute of Santiago de Compostela (IDIS), Spain
| | - Irene Zarra-Ferro
- Pharmacy Department, University Clinical Hospital Santiago de Compostela (CHUS), Spain; Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Spain
| | - Dolors Soy
- Pharmacy Department Division of Medicines, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain; Department of Pharmacology, Toxicology and Chemical Therapeutics, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Anxo Fernández-Ferreiro
- Pharmacy Department, University Clinical Hospital Santiago de Compostela (CHUS), Spain; Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Spain.
| | - Víctor Mangas-Sanjuan
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Valencia, Spain; Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia - University of Valencia, Valencia, Spain.
| | - Cristina Mondelo-García
- Pharmacy Department, University Clinical Hospital Santiago de Compostela (CHUS), Spain; Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Spain.
| |
Collapse
|
6
|
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
|
7
|
Anhøj S, Ebdrup B, Nielsen MØ, Antonsen P, Glenthøj B, Rostrup E. Functional Connectivity Between Auditory and Medial Temporal Lobe Networks in Antipsychotic-Naïve Patients With First-Episode Schizophrenia Predicts the Effects of Dopamine Antagonism on Auditory Verbal Hallucinations. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:308-316. [PMID: 38298804 PMCID: PMC10829637 DOI: 10.1016/j.bpsgos.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 02/02/2024] Open
Abstract
Background Understanding how antipsychotic medication ameliorates auditory verbal hallucinations (AVHs) through modulation of brain circuitry is pivotal for understanding the pathophysiology of psychosis and for predicting treatment response. Methods This case-control study included examinations at baseline and at follow-up after 6 weeks. Initially, antipsychotic-naïve patients with first-episode schizophrenia who were experiencing AVHs were recruited together with healthy control participants. Antipsychotic treatment with the relatively selective D2 receptor antagonist amisulpride was administered as monotherapy. Functional connectivity measured by resting-state functional magnetic resonance imaging between networks of interest was used to study the effects of D2 blockade on brain circuitry and predict clinical treatment response. Hallucinations were rated with the Positive and Negative Syndrome Scale. Results Thirty-two patients experiencing AVHs and 34 healthy control participants were scanned at baseline. Twenty-two patients and 34 healthy control participants were rescanned at follow-up. Connectivity between the auditory network and the medial temporal lobe network was increased in patients at baseline (p = .002) and normalized within 6 weeks of D2 blockade (p = .018). At baseline, the connectivity between these networks was positively correlated with ratings of hallucinations (t = 2.67, p = .013). Moreover, baseline connectivity between the auditory network and the medial temporal lobe network predicted reduction in hallucinations (t = 2.34, p = .032). Conclusions Functional connectivity between the auditory network and the medial temporal lobe predicted response to initial antipsychotic treatment. These findings demonstrate that connectivity between networks involved in auditory processing, internal monitoring, and memory is associated with the clinical effect of dopamine antagonism.
Collapse
Affiliation(s)
- Simon Anhøj
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, Glostrup, Denmark
| | - Bjørn Ebdrup
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Ødegaard Nielsen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patrick Antonsen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, Glostrup, Denmark
| | - Birte Glenthøj
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Egill Rostrup
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center Glostrup, Glostrup, Denmark
| |
Collapse
|
8
|
Yamasaki F, Kanahara N, Nakata Y, Koyoshi S, Yanagisawa Y, Saito T, Oiwa T, Kogure M, Sasaki T, Yoshida T, Kimura H, Iyo M. Can brexpiprazole be switched safely in patients with schizophrenia and dopamine supersensitivity psychosis? A retrospective analysis in a real-world clinical practice. J Psychopharmacol 2023; 37:992-1002. [PMID: 37395368 DOI: 10.1177/02698811231177268] [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] [Indexed: 07/04/2023]
Abstract
BACKGROUND Several studies have reported that a switch to the dopamine partial agonist (DPA) aripiprazole (ARP), especially when the switch is abrupt, is likely to fail and sometimes worsen psychosis in schizophrenia patients already under high-dose antipsychotic treatment. Such a switching failure is speculated to be related to be the dopamine supersensitivity state. The risks of switching to the DPA brexpiprazole (BREX) have not been reported. AIMS AND METHODS We retrospectively analyzed the cases of 106 patients with schizophrenia to identify any factors related to the success or failure of switching to BREX. RESULTS The comparison between the patients with dopamine supersensitivity psychosis (n = 44) and those without (n = 62) revealed no significant difference in the switching failure judged at the sixth week. A comparison of the patients with successful switching (n = 80) and those who failed (n = 26) revealed that patients with treatment-resistant schizophrenia (TRS) were significantly more likely to fail. A logistic regression analysis also revealed that patients with past failure of switching to ARP are likely to succeed in switching to BREX. The 2-year follow-up of the patients with successful switching to BREX suggested that the patients who were treated with BREX, even temporarily, experienced some improvement in their Global Assessment of Functioning and Clinical Global Impression-Severity scores. CONCLUSIONS Overall, the results indicate that patients with schizophrenia can be switched more safely to BREX compared to ARP. However, the failure of switching to BREX could be higher in patients with TRS, and thus, starting BREX treatment in refractory patients warrants careful monitoring.
Collapse
Affiliation(s)
- Fumiaki Yamasaki
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Psychiatry, Douwa-kai Chiba Hospital, Funabashi, Japan
| | - Nobuhisa Kanahara
- Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, Chiba, Japan
- Department of Psychiatry, Shirayuri-kai Ichihara Tsuruoka Hospital, Ichihara, Japan
| | - Yusuke Nakata
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinji Koyoshi
- Department of Psychiatry, Doujin-kai Kisarazu Hospital, Kisarazu, Japan
| | - Yuta Yanagisawa
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Psychiatry, Gakuji-kai Kimura Hospital, Chiba, Japan
| | - Takeru Saito
- Department of Psychiatry, Naoki-kai Isogaya Hospital, Ichihara, Japan
| | - Takahiro Oiwa
- Department of Psychiatry, Naoki-kai Isogaya Hospital, Ichihara, Japan
| | - Masanobu Kogure
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tsuyoshi Sasaki
- Department of Psychiatry, Douwa-kai Chiba Hospital, Funabashi, Japan
- Department of Child Psychiatry, Chiba University Hospital, Chiba, Japan
| | - Taisuke Yoshida
- Department of Psychiatry, Doujin-kai Kisarazu Hospital, Kisarazu, Japan
| | - Hiroshi Kimura
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Psychiatry, Gakuji-kai Kimura Hospital, Chiba, Japan
- Department of Psychiatry, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
9
|
O’Neill JR, Taylor DM, Horowitz MA. Implementing gradual, hyperbolic tapering of long-acting injectable antipsychotics by prolonging the inter-dose interval: an in silico modelling study. Ther Adv Psychopharmacol 2023; 13:20451253231198463. [PMID: 37719449 PMCID: PMC10501077 DOI: 10.1177/20451253231198463] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 08/08/2023] [Indexed: 09/19/2023] Open
Abstract
Gradual, hyperbolic tapering has been proposed as a method to reduce the risk of withdrawal effects and potential relapse of an underlying condition by minimising disruption of existing equilibria. We applied hyperbolic tapering principles in silico to long-acting aripiprazole to generate regimens for withdrawal in clinical practice. We derived thresholds for taper rates using existing studies and consensus. Using pharmacokinetic data for aripiprazole long-acting injectable antipsychotic (ALAI), we conducted in silico modelling to examine the impact of abrupt cessation of long-acting injectable antipsychotic (LAI) medication and the effect of prolonging inter-dose interval on plasma aripiprazole levels and consequent D2 occupancy. We also modelled transitions from LAI medication to oral medication. Regimens were designed to afford a rate of reduction between 5 and 12.5 percentage points of D2 occupancy per month. Abrupt discontinuation of ALAI was shown to lead to a maximal D2 occupancy reduction of 16.8 percentage points per month; prolongation of the inter-dose interval of ALAI produced a slower reduction. Specifically, hyperbolic tapering was afforded by prolongation of a 400 mg ALAI inter-dose interval from 4 to 7 weeks, before reducing the dose to 300 mg ALAI. This could then be administered at up to 4-week (for 6% maximal D2 occupancy change), 6-week (9% change) or 7-week (11% change) intervals. Switching to oral medication - 5, 2.5 and 1.25 mg for the three regimens, respectively - is required for ALAI to complete full cessation to prevent too rapid a reduction in D2 occupancy. Oral medication should probably be maintained at a consistent dose for 3-6 months before further reductions to account for residual LAI being concurrently eliminated. Hyperbolic dose tapering is possible with ALAI through prolongation of the inter-dose interval and may reduce the risk of relapse compared to abrupt discontinuation of LAI medication.
Collapse
Affiliation(s)
- James R. O’Neill
- South West Yorkshire Partnership NHS Foundation Trust, Newton Lodge, Ouchthorpe Lane, Wakefield WF1 3SP, UK
| | | | | |
Collapse
|
10
|
Siafis S, Wu H, Wang D, Burschinski A, Nomura N, Takeuchi H, Schneider-Thoma J, Davis JM, Leucht S. Antipsychotic dose, dopamine D2 receptor occupancy and extrapyramidal side-effects: a systematic review and dose-response meta-analysis. Mol Psychiatry 2023; 28:3267-3277. [PMID: 37537284 PMCID: PMC10618092 DOI: 10.1038/s41380-023-02203-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 07/17/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023]
Abstract
Antipsychotic drugs differ in their propensity to cause extrapyramidal side-effects (EPS), but their dose-effects are unclear. Therefore, we conducted a systematic review and dose-response meta-analysis. We searched multiple electronic databases up to 20.02.2023 for fixed-dose studies investigating 16 second-generation antipsychotics and haloperidol (all formulations and administration routes) in adults with acute exacerbations of schizophrenia. The primary outcome was the number of participants receiving antiparkinsonian medication, and if not available, the number of participants with extrapyramidal side-effects (EPS) and the mean scores of EPS rating scales were used as proxies. The effect-size was odds ratio (ORs) compared with placebo. One-stage random-effects dose-response meta-analyses with restricted cubic splines were conducted to estimate the dose-response curves. We also examined the relationship between dopamine D2 receptor (D2R) occupancy and ORs by estimating occupancies from administrated doses. We included data from 110 studies with 382 dose arms (37193 participants). Most studies were short-term with median duration of 6 weeks (range 3-26 weeks). Almost all antipsychotics were associated with dose-dependent EPS with varied degrees and the maximum ORs ranged from OR = 1.57 95%CI [0.97, 2.56] for aripiprazole to OR = 7.56 95%CI [3.16, 18.08] for haloperidol at 30 mg/d. Exceptions were quetiapine and sertindole with negligible risks across all doses. There was very low quality of findings for cariprazine, iloperidone, and zotepine, and no data for clozapine. The D2R occupancy curves showed that the risk increased substantially when D2R occupancy exceeded 75-85%, except for D2R partial agonists that had smaller ORs albeit high D2R occupancies. In conclusion, we found that the risk of EPS increases with rising doses and differs substantially in magnitude among antipsychotics, yet exceptions were quetiapine and sertindole with negligible risks. Our data provided additional insights into the current D2R therapeutic window for EPS.
Collapse
Affiliation(s)
- Spyridon Siafis
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany.
| | - Hui Wu
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Dongfang Wang
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Angelika Burschinski
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nobuyuki Nomura
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyoshi Takeuchi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Johannes Schneider-Thoma
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - John M Davis
- Psychiatric Institute, University of Illinois at Chicago, Chicago, IL, USA
- Maryland Psychiatric Research Center, Baltimore, MD, USA
| | - Stefan Leucht
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
11
|
González-Rodríguez A, Monreal JA, Natividad M, Seeman MV. Seventy Years of Treating Delusional Disorder with Antipsychotics: A Historical Perspective. Biomedicines 2022; 10:biomedicines10123281. [PMID: 36552037 PMCID: PMC9775530 DOI: 10.3390/biomedicines10123281] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
For many decades, delusional disorder (DD) has been considered a treatment-resistant disorder, with antipsychotics acknowledged as the best, though imperfect, treatment. It is possible that the discovery of the right drug could turn treatment resistance into treatment response. The goal of this narrative review is to provide a historical perspective of the treatment of DD since the introduction of antipsychotics 70 years ago. The following search terms were used to scan the literature: antipsychotics AND "delusional disorder". Findings were that therapy for DD symptoms has changed over time. Initial reports suggested that the drug of choice was the antipsychotic pimozide, and that this drug was especially effective for the somatic subtype of DD. Subsequent studies demonstrated that other antipsychotics, for instance, risperidone and olanzapine, were also highly effective. Treatment response may vary according to the presence or absence of specific symptoms, such as cognitive defect and depression. Clozapine, partial D2 agonists, and long-acting injectable drugs may be more effective than other drugs, but the evidence is not yet in. Because of the absence of robust evidence, treatment guidelines for the optimal management of DD are not yet available.
Collapse
Affiliation(s)
- Alexandre González-Rodríguez
- Department of Mental Health, Mutua Terrassa University Hospital, Fundació Docència I Recerca Mutua Terrassa, University of Barcelona (UB), CIBERSAM, 5 Dr Robert Square, 08221 Terrassa, Spain
- Correspondence:
| | - José A. Monreal
- Department of Mental Health, Mutua Terrassa University Hospital, Fundació Docència I Recerca Mutua Terrassa, University of Barcelona (UB), CIBERSAM, 5 Dr Robert Square, 08221 Terrassa, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), 08221 Terrassa, Spain
| | - Mentxu Natividad
- Department of Mental Health, Mutua Terrassa University Hospital, Fundació Docència I Recerca Mutua Terrassa, University of Barcelona (UB), CIBERSAM, 5 Dr Robert Square, 08221 Terrassa, Spain
| | - Mary V. Seeman
- Department of Psychiatry, University of Toronto, 605 260 Health Street West, Toronto, ON M5P 3L6, Canada
| |
Collapse
|
12
|
Ueda N, Tanaka K, Maruo K, Roach N, Sumiyoshi T, Watanabe K, Hanakawa T. Perceptual inference, accuracy, and precision in temporal reproduction in schizophrenia. Schizophr Res Cogn 2022; 28:100229. [PMID: 34976749 PMCID: PMC8683762 DOI: 10.1016/j.scog.2021.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022]
Abstract
Accumulating evidence suggests that deficits in perceptual inference account for symptoms of schizophrenia. One manifestation of perceptual inference is the central bias, i.e., the tendency to put emphasis on prior experiences over actual events in perceiving incoming sensory stimuli. Using an interval reproduction task, this study aimed to determine whether patients with schizophrenia show a stronger central bias than participants without schizophrenia. In the interval reproduction task, participants were shown a cross on a screen. The cross was replaced with a Gaussian patch for a predetermined time interval, and participants were required to reproduce the interval duration by pressing and releasing the space key. We manipulated the uncertainty of prior information using different interval distributions. We found no difference in the influence of prior information on interval reproduction between patients and controls. However, patients with SZ showed a stronger central bias than healthy participants in the intermediate interval range (approximately 450 ms to 900 ms). It is possible that the patients in SZ have non-uniform deficits associated with interval range or uncertainty of prior information in perceptual inference. Further, the severity of avolition and alogia was correlated with the strength of central bias in SZ. This study provides some insights into the mechanisms underlying the association between schizophrenic symptoms and perceptual inference.
Collapse
Affiliation(s)
- Natsuki Ueda
- NCNP Brain Physiology and Pathology, Tokyo Medical and Dental University Graduate School, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8551, Japan
- Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8551, Japan
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8551, Japan
| | - Kanji Tanaka
- Faculty of Arts and Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Waseda Institute for Advanced Study, Waseda University, 1-21-1 Nishi Waseda, Shinjuku-ku, Tokyo 169-0051, Japan
| | - Kazushi Maruo
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Neil Roach
- School of Psychology, University of Nottingham, NG7 2RD Nottingham, United Kingdom
| | - Tomiki Sumiyoshi
- Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8551, Japan
| | - Katsumi Watanabe
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Faculty of Arts, Design, and Architecture, University of New South Wales, Cnr Oxford St & Greens Rd, Paddington, NSW 2021, Australia
| | - Takashi Hanakawa
- NCNP Brain Physiology and Pathology, Tokyo Medical and Dental University Graduate School, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8551, Japan
- Integrated Neuroanatomy and Neuroimaging, Kyoto University Graduate School of Medicine, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
13
|
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
|
14
|
Kiss B, Krámos B, Laszlovszky I. Potential Mechanisms for Why Not All Antipsychotics Are Able to Occupy Dopamine D 3 Receptors in the Brain in vivo. Front Psychiatry 2022; 13:785592. [PMID: 35401257 PMCID: PMC8987915 DOI: 10.3389/fpsyt.2022.785592] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/25/2022] [Indexed: 11/29/2022] Open
Abstract
Dysfunctions of the dopaminergic system are believed to play a major role in the core symptoms of schizophrenia such as positive, negative, and cognitive symptoms. The first line of treatment of schizophrenia are antipsychotics, a class of medications that targets several neurotransmitter receptors in the brain, including dopaminergic, serotonergic, adrenergic and/or muscarinic receptors, depending on the given agent. Although the currently used antipsychotics display in vitro activity at several receptors, majority of them share the common property of having high/moderate in vitro affinity for dopamine D2 receptors (D2Rs) and D3 receptors (D3Rs). In terms of mode of action, these antipsychotics are either antagonist or partial agonist at the above-mentioned receptors. Although D2Rs and D3Rs possess high degree of homology in their molecular structure, have common signaling pathways and similar in vitro pharmacology, they have different in vivo pharmacology and therefore behavioral roles. The aim of this review, with summarizing preclinical and clinical evidence is to demonstrate that while currently used antipsychotics display substantial in vitro affinity for both D3Rs and D2Rs, only very few can significantly occupy D3Rs in vivo. The relative importance of the level of endogenous extracellular dopamine in the brain and the degree of in vitro D3Rs receptor affinity and selectivity as determinant factors for in vivo D3Rs occupancy by antipsychotics, are also discussed.
Collapse
Affiliation(s)
- Béla Kiss
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Balázs Krámos
- Spectroscopic Research Department, Gedeon Richter Plc., Budapest, Hungary
| | | |
Collapse
|
15
|
Remington G, Hahn MK, Agarwal SM, Chintoh A, Agid O. Schizophrenia: Antipsychotics and drug development. Behav Brain Res 2021; 414:113507. [PMID: 34352293 DOI: 10.1016/j.bbr.2021.113507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022]
Abstract
The introduction of chlorpromazine and the work that ensued provided the foundation to reposition schizophrenia as a biological illness. The present paper follows the evolution of antipsychotics and their shift from 'typical' to 'atypical'. Atypicality is reviewed in reference to its original definition, clozapine's role, and developments that now leave the concept's utility in question. In a similar fashion, drug development is reviewed in the context of the illness' multiple symptom domains, as well as differences captured by clinical staging and phenotyping. Collectively, the evidence argues for a more nuanced approach to drug development that aligns with the illness' heterogeneity and complexity. Just as 'atypical' as a descriptor for antipsychotics may be outdated, it may be time to set aside the notion of developing drugs that treat 'schizophrenia'.
Collapse
Affiliation(s)
- Gary Remington
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada.
| | - Margaret K Hahn
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Sri Mahavir Agarwal
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Araba Chintoh
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Ofer Agid
- University of Toronto, Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| |
Collapse
|
16
|
Smart K, Naganawa M, Baldassarri SR, Nabulsi N, Ropchan J, Najafzadeh S, Gao H, Navarro A, Barth V, Esterlis I, Cosgrove KP, Huang Y, Carson RE, Hillmer AT. PET Imaging Estimates of Regional Acetylcholine Concentration Variation in Living Human Brain. Cereb Cortex 2021; 31:2787-2798. [PMID: 33442731 PMCID: PMC8355478 DOI: 10.1093/cercor/bhaa387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/06/2020] [Accepted: 12/01/2020] [Indexed: 11/13/2022] Open
Abstract
Acetylcholine (ACh) has distinct functional roles in striatum compared with cortex, and imbalance between these systems may contribute to neuropsychiatric disease. Preclinical studies indicate markedly higher ACh concentrations in the striatum. The goal of this work was to leverage positron emission tomography (PET) imaging estimates of drug occupancy at cholinergic receptors to explore ACh variation across the human brain, because these measures can be influenced by competition with endogenous neurotransmitter. PET scans were analyzed from healthy human volunteers (n = 4) and nonhuman primates (n = 2) scanned with the M1-selective radiotracer [11C]LSN3172176 in the presence of muscarinic antagonist scopolamine, and human volunteers (n = 10) scanned with the α4β2* nicotinic ligand (-)-[18F]flubatine during nicotine challenge. In all cases, occupancy estimates within striatal regions were consistently lower (M1/scopolamine human scans, 31 ± 3.4% occupancy in striatum, 43 ± 2.9% in extrastriatal regions, p = 0.0094; nonhuman primate scans, 42 ± 26% vs. 69 ± 28%, p < 0.0001; α4β2*/nicotine scans, 67 ± 15% vs. 74 ± 16%, p = 0.0065), indicating higher striatal ACh concentration. Subject-level measures of these concentration differences were estimated, and whole-brain images of regional ACh concentration gradients were generated. These results constitute the first in vivo estimates of regional variation in ACh concentration in the living brain and offer a novel experimental method to assess potential ACh imbalances in clinical populations.
Collapse
Affiliation(s)
- Kelly Smart
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mika Naganawa
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
| | - Stephen R Baldassarri
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nabeel Nabulsi
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jim Ropchan
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Hong Gao
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
| | | | | | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, USA
| | - Kelly P Cosgrove
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, USA
| | - Yiyun Huang
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
| | - Richard E Carson
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Ansel T Hillmer
- Yale PET Center, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
17
|
Kotzalidis GD, Rapinesi C, Chetoni C, De Filippis S. Aripiprazole IM depot as an option for the treatment of bipolar disorder. Expert Opin Pharmacother 2021; 22:1407-1416. [PMID: 33847183 DOI: 10.1080/14656566.2021.1910236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Introduction: Long-acting injectable (LAI) antipsychotic drugs are developed to reduce daily intake need and to overcome treatment non-adherence. Aripiprazole IM depot refers to two long-acting aripiprazole formulations, once monthly monohydrate (AOM) and aripiprazole lauroxil. AOM has been approved for schizophrenia since 2012 and for bipolar disorder since 2017. Aripiprazole lauroxil is approved for schizophrenia, not for bipolar disorder.Areas covered: To assess the effect of AOM in bipolar disorder, the authors searched PubMed and ClinicalTrials.gov for randomized trials using AOM in patients with bipolar disorder. Included were four studies covering efficacy, functioning, quality of life, and safety/tolerability. Studies lasted 12 months.Expert opinion: AOM reduced symptoms of patients with bipolar disorder and a manic episode, increased functioning and quality of life, and protected from recurrence of manic episodes. It proved to be safe/tolerable, with only akathisia occurring in ≥10% of cases and more frequently than with placebo. However, there were only 143 patients receiving AOM in the considered studies. Included studies were backed in their conclusions by other literature, but they come from 2017-2018. No studies are expected or planned in the near future. Aripiprazole lauroxil has not applied for approval in bipolar disorder and there is no sign it will.
Collapse
Affiliation(s)
- Georgios D Kotzalidis
- NESMOS Department, Sapienza University of Rome, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Rome, Italy.,Department of Neuropsychiatry, Villa Von Siebenthal Neuropsychiatric Hospital, Genzano Di Roma, Italy
| | - Chiara Rapinesi
- NESMOS Department, Sapienza University of Rome, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Rome, Italy
| | - Chiara Chetoni
- NESMOS Department, Sapienza University of Rome, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Rome, Italy
| | - Sergio De Filippis
- Department of Neuropsychiatry, Villa Von Siebenthal Neuropsychiatric Hospital, Genzano Di Roma, Italy
| |
Collapse
|
18
|
Kimura H, Kanahara N, Iyo M. Rationale and neurobiological effects of treatment with antipsychotics in patients with chronic schizophrenia considering dopamine supersensitivity. Behav Brain Res 2021; 403:113126. [PMID: 33460681 DOI: 10.1016/j.bbr.2021.113126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
The long-term treatment of patients with schizophrenia often involves the management of relapses for most patients and the development of treatment resistance in some patients. To stabilize the clinical course and allow as many patients as possible to recover, clinicians need to recognize dopamine supersensitivity, which can be provoked by administration of high dosages of antipsychotics, and deal with it properly. However, no treatment guidelines have addressed this issue. The present review summarized the characteristics of long-acting injectable antipsychotics, dopamine partial agonists, and clozapine in relation to dopamine supersensitivity from the viewpoints of receptor profiles and pharmacokinetics. The potential merits and limitations of these medicines are discussed, as well as the risks of treating patients with established dopamine supersensitivity with these classes of drugs. Finally, the review discussed the biological influence of antipsychotic treatment on the human brain based on findings regarding the relationship between the hippocampus and antipsychotics.
Collapse
Affiliation(s)
- Hiroshi Kimura
- Department of Psychiatry, School of Medicine, International University of Health and Welfare, Chiba, Japan; Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan; Department of Psychiatry, Gakuji-kai Kimura Hospital, Chiba, Japan.
| | - Nobuhisa Kanahara
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan; Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
19
|
Carvalho Henriques B, Yang EH, Lapetina D, Carr MS, Yavorskyy V, Hague J, Aitchison KJ. How Can Drug Metabolism and Transporter Genetics Inform Psychotropic Prescribing? Front Genet 2020; 11:491895. [PMID: 33363564 PMCID: PMC7753050 DOI: 10.3389/fgene.2020.491895] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/25/2020] [Indexed: 12/11/2022] Open
Abstract
Many genetic variants in drug metabolizing enzymes and transporters have been shown to be relevant for treating psychiatric disorders. Associations are strong enough to feature on drug labels and for prescribing guidelines based on such data. A range of commercial tests are available; however, there is variability in included genetic variants, methodology, and interpretation. We herein provide relevant background for understanding clinical associations with specific variants, other factors that are relevant to consider when interpreting such data (such as age, gender, drug-drug interactions), and summarize the data relevant to clinical utility of pharmacogenetic testing in psychiatry and the available prescribing guidelines. We also highlight areas for future research focus in this field.
Collapse
Affiliation(s)
| | - Esther H. Yang
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Diego Lapetina
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Michael S. Carr
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Vasyl Yavorskyy
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Joshua Hague
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Katherine J. Aitchison
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Herms EN, Bishop JR, Okuneye VT, Tamminga CA, Keshavan MS, Pearlson GD, Clementz BA, McDowell JE, Ivleva EI, Gershon ES, Sweeney JA, Keedy SK. No connectivity alterations for striatum, default mode, or salience network in association with self-reported antipsychotic medication dose in a large chronic patient group. Schizophr Res 2020; 223:359-360. [PMID: 32624351 PMCID: PMC8082971 DOI: 10.1016/j.schres.2020.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/01/2020] [Accepted: 06/18/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Emma N. Herms
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - Jeffrey R. Bishop
- Departments of Pharmacy and Psychiatry, University of Minnesota, Minneapolis, MN, United States
| | - Victoria T. Okuneye
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - Carol A. Tamminga
- Department of Psychiatry, UT-Southwestern Medical Center, Dallas, TX, United States
| | - Matcheri S. Keshavan
- Department of Psychiatry, Beth Israel Deaconness Medical Center and Harvard Medical School, Boston, MA, United States
| | - Godfrey D. Pearlson
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, CT, United States; Institute of Living, Hartford Hospital, Hartford, CT, United States
| | - Brett A. Clementz
- Department of Psychology and Neuroscience, University of Georgia, Athens, GA, United States
| | - Jennifer E. McDowell
- Department of Psychology and Neuroscience, University of Georgia, Athens, GA, United States
| | - Elena I. Ivleva
- Department of Psychiatry, UT-Southwestern Medical Center, Dallas, TX, United States
| | - Elliot S. Gershon
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - John A. Sweeney
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, United States
| | - Sarah K. Keedy
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| |
Collapse
|
21
|
Tsartsalis S, Tournier BB, Millet P. In vivo absolute quantification of striatal and extrastriatal D 2/3 receptors with [ 123I]epidepride SPECT. EJNMMI Res 2020; 10:66. [PMID: 32548681 PMCID: PMC7297889 DOI: 10.1186/s13550-020-00650-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/27/2020] [Indexed: 01/08/2023] Open
Abstract
Background [123I]epidepride is a high-affinity radiotracer used in single-photon emission computed tomography (SPECT) imaging of the D2/3 receptors. It binds with high affinity to striatal and extrastriatal receptors. Nevertheless, its slow kinetics in the striatum impedes quantification in this region. Thus, an approach that would allow a simultaneous quantification of both striatal and extrastriatal D2/3 receptors would be of interest for preclinical and clinical SPECT neuroimaging. We describe a partial saturation protocol that allows us to produce an in vivo Scatchard plot and thus estimate Bavail and appKd separately in both striatal and extrastriatal regions, through a single dynamic SPECT session. To validate this approach, a multi-injection protocol is used for the full kinetic modeling of [123I]epidepride using a two-tissue compartment, 5-parameter model (2T-5k). Methods Eighteen male rats were used. Binding parameters were estimated using the multi-injection protocol. Various simulations were performed to estimate the optimal conditions for the partial saturation protocol, which was applied at the region and voxel level. The results of the partial saturation study were compared to those obtained with the 2T-5k model. To illustrate the interest of the partial saturation approach, we performed a preliminary study of the effect of a chronic, subcutaneous administration of haloperidol (1 mg/kg/day), a D2 receptor antagonist, on the Bavail of [123I]epidepride in the rat striatum. Results A series of simulations demonstrated that a mass of 3 ug/kg of unlabeled epidepride allows the formation of an in vivo Scatchard plot. The partial saturation study led to robust estimations of Bavail in all brain regions that highly correlated (r = 0.99) with the corresponding values from the multi-injection study. A chronic haloperidol treatment resulted in a 17.9% increase in the Bavail values in the left Caudate Putamen nucleus (CP) (p = 0.07) and a 13.8% increase in the right CP (p = 0.12). Conclusion A partial saturation method allowed the robust quantification of D2/3 receptors in striatal and extrastriatal D2/3 receptors with a single-scan approach. This approach may be applied in the mapping of the D2/3 receptor in translational biological studies and potentially, in clinical SPECT imaging.
Collapse
Affiliation(s)
- Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1226, Thônex, Switzerland.,Division of Psychiatric Specialties, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1226, Thônex, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1226, Thônex, Switzerland. .,Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
22
|
Girgis RR, Forbes A, Abi-Dargham A, Slifstein M. A positron emission tomography occupancy study of brexpiprazole at dopamine D 2 and D 3 and serotonin 5-HT 1A and 5-HT 2A receptors, and serotonin reuptake transporters in subjects with schizophrenia. Neuropsychopharmacology 2020; 45:786-792. [PMID: 31847007 PMCID: PMC7075883 DOI: 10.1038/s41386-019-0590-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/20/2019] [Accepted: 12/05/2019] [Indexed: 11/09/2022]
Abstract
The objective of this study (NCT01854944) was to assess D2/D3, 5-HT1A, 5-HT2A and serotonin transporter (SERT) occupancies of brexpiprazole in adult subjects with schizophrenia in order to identify the in vivo pharmacologic profile that may be relevant to the antipsychotic, antidepressant, and side effect profiles of the drug. Subjects were grouped into three independent cohorts of four subjects each. All subjects underwent positron emission tomography (PET) scans with two different radiotracers at baseline prior to brexpiprazole administration, and again on Day 10 after daily doses of either 4 mg (Cohorts 1 and 2), or 1 mg (Cohort 3). Cohort 1 received scans with [11C]-(+)-PHNO to measure D2 and D3 receptor occupancy and [11C]CUMI101 to measure 5-HT1A occupancy; Cohort 2 received [11C]MDL100907 for 5-HT2A occupancy and [11C]DASB for SERT occupancy; Cohort 3 underwent scanning with [11C]-(+)-PHNO and [11C]MDL100907. Five female and seven male subjects, aged 42 ± 8 years (range, 28-55 years), participated in this study. Dose dependency was observed at D2 receptors, with occupancies reaching 64 ± 8% (mean +/- SD) following 1 mg/day and 80 ± 12% following 4 mg/day. D3 receptor availability increased following 1 mg brexpiprazole treatment and did not change with 4 mg. Robust and dose-related occupancy was also observed at 5-HT2A receptors. Negligible occupancy (<5%) was observed at 5-HT1A and SERT at 4 mg/day. In summary, brexpiprazole demonstrated in vivo binding to D2 receptors and 5-HT2A receptors at steady state after 10 days of daily administration in a dose dependent manner, while binding to D3, 5-HT1A receptors and SERT was not detectable with the radiotracers used for these targets. This pharmacologic profile is consistent with the observed antipsychotic and antidepressant effects.
Collapse
Affiliation(s)
- Ragy R Girgis
- New York State Psychiatric Institute, Columbia University Irving Medical Center, New York, NY, USA.
| | - Andy Forbes
- Otsuka Pharmaceutical Development & Commercialization Inc., Princeton, NJ, USA
| | - Anissa Abi-Dargham
- Department of Psychiatry, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Mark Slifstein
- Department of Psychiatry, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
23
|
In response to the letter “[11C]raclopride and extrastriatal binding to D2/3 receptors”. Neuroimage 2020; 207:116371. [DOI: 10.1016/j.neuroimage.2019.116371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/16/2019] [Indexed: 11/23/2022] Open
|
24
|
Obayashi Y, Mitsui S, Sakamoto S, Minao N, Yoshimura B, Kono T, Yada Y, Okahisa Y, Takao S, Kishi Y, Takeda T, Takaki M, Yamada N. Switching strategies for antipsychotic monotherapy in schizophrenia: a multi-center cohort study of aripiprazole. Psychopharmacology (Berl) 2020; 237:167-175. [PMID: 31624859 DOI: 10.1007/s00213-019-05352-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022]
Abstract
RATIONALE Changing antipsychotics of patients with chronic schizophrenia involves several risks. Switching to aripiprazole is especially difficult. We investigated switching methods and related factors for successful switching patients with chronic schizophrenia to aripiprazole. OBJECTIVES This study was a multi-center historical cohort study and approved by the research ethics committee of Okayama University Hospital and Okayama Psychiatric Medical Center. We compared survival proportions of 178 chronic schizophrenia patients who continued aripiprazole monotherapy for 6 months after non-direct switching (add-on switching (n = 45), cross switching (n = 62)) or direct switching (n = 71). We adjusted possible confounders using a Cox proportional hazards model. RESULTS Of patients with chronic schizophrenia, 56.7% (101/178) were switched to aripiprazole monotherapy, and 55.0% (98/178) showed improvement in symptoms as demonstrated by the Clinical Global Impression Severity score. Kaplan-Meier survival curves showed that non-direct switching had a higher survival proportion than direct switching (log-rank test, p = 0.012). Even after adjusting for several variables using a Cox proportional hazards model, add-on switching had a significantly lower hazard at 6 months than direct switching (hazard ratio 0.42, 95% confidence interval 0.21-0.82, P = 0.01). In cases of switching to aripiprazole for psychiatric symptoms, non-direct switching had a lower hazard than direct switching (hazard ratio 0.41, 95% confidence interval 0.21-0.81, P = 0.01) but was not significant for adverse reaction. When aripiprazole was switched from olanzapine, add-on switch showed the lowest hazard ratio for continuation (hazard ratio 0.29, 95% confidence interval 0.07-1.11, P = 0.07). CONCLUSIONS Flexibility in strategies when switching to aripiprazole may induce a better outcome for patients with chronic schizophrenia.
Collapse
Affiliation(s)
- Yoshiaki Obayashi
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Psychiatry, Okayama Psychiatric Medical Center, Okayama, Japan
- Department of Psychiatry, Zikei Hospital, Okayama, Japan
- Department of Psychiatry, Fukuyama Kokorono Hospital, Fukuyama, Japan
| | - Satoshi Mitsui
- Department of Epidemiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Sakamoto
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nozomu Minao
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Bunta Yoshimura
- Department of Psychiatry, Okayama Psychiatric Medical Center, Okayama, Japan
| | - Toshiki Kono
- Department of Psychiatry, Okayama Psychiatric Medical Center, Okayama, Japan
| | - Yuji Yada
- Department of Psychiatry, Okayama Psychiatric Medical Center, Okayama, Japan
| | - Yuko Okahisa
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Soshi Takao
- Department of Epidemiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshiki Kishi
- Department of Psychiatry, Okayama Psychiatric Medical Center, Okayama, Japan
| | | | - Manabu Takaki
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Norihito Yamada
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
25
|
Whitton AE, Green AI, Pizzagalli DA, Roth RM, Williams JM, Brunette MF. Potent Dopamine D2 Antagonists Block the Reward-Enhancing Effects of Nicotine in Smokers With Schizophrenia. Schizophr Bull 2019; 45:1300-1308. [PMID: 30690638 PMCID: PMC6811816 DOI: 10.1093/schbul/sby185] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Antipsychotics that are potent dopamine (DA) D2 receptor antagonists have been linked to elevated levels of nicotine dependence in smokers with schizophrenia. Because activation of D2 receptors mediates motivation for nicotine, we examined whether potent D2 antagonists would diminish nicotine's ability to stimulate reward processing-a mechanism that may drive compensatory increases in smoking. Smokers with schizophrenia (n = 184) were recruited and stratified into medication groups based on D2 receptor antagonist potency. The effects of smoking on reward function were assessed using a probabilistic reward task (PRT), administered pre- and post-smoking. The PRT used an asymmetrical reinforcement schedule to produce a behavioral response bias, previously found to increase under conditions (including smoking) that enhance mesolimbic DA signaling. Among the 98 participants with valid PRT data and pharmacotherapy that could be stratified into D2 receptor antagonism potency, a medication × smoking × block interaction emerged (P = .005). Post-hoc tests revealed a smoking × block interaction only for those not taking potent D2 antagonists (P = .007). This group exhibited smoking-related increases in response bias (P < .001) that were absent in those taking potent D2 antagonists (P > .05). Our findings suggest that potent D2 antagonists diminish the reward-enhancing effects of nicotine in smokers with schizophrenia. This may be a mechanism implicated in the increased rate of smoking often observed in patients prescribed these medications. These findings have important clinical implications for the treatment of nicotine dependence in schizophrenia.
Collapse
Affiliation(s)
- Alexis E Whitton
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Belmont, MA,Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Alan I Green
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, NH,Dartmouth Clinical and Translational Science Institute, Hanover, NH
| | - Diego A Pizzagalli
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Belmont, MA,Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Robert M Roth
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Jill M Williams
- Department of Psychiatry, Rutgers University, New Brunswick, NJ
| | - Mary F Brunette
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, NH,To whom correspondence should be addressed; Department of Psychiatry, Dartmouth-Hitchcock, 2 Pillsbury Street, Suite 40, Concord, NH 03301, US; tel: 603-229-5419, fax: 603-271-5262, e-mail:
| |
Collapse
|
26
|
Kaar SJ, Natesan S, McCutcheon R, Howes OD. Antipsychotics: Mechanisms underlying clinical response and side-effects and novel treatment approaches based on pathophysiology. Neuropharmacology 2019; 172:107704. [PMID: 31299229 DOI: 10.1016/j.neuropharm.2019.107704] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/13/2019] [Accepted: 07/08/2019] [Indexed: 12/17/2022]
Abstract
Antipsychotic drugs are central to the treatment of schizophrenia and other psychotic disorders but are ineffective for some patients and associated with side-effects and nonadherence in others. We review the in vitro, pre-clinical, clinical and molecular imaging evidence on the mode of action of antipsychotics and their side-effects. This identifies the key role of striatal dopamine D2 receptor blockade for clinical response, but also for endocrine and motor side-effects, indicating a therapeutic window for D2 blockade. We consider how partial D2/3 receptor agonists fit within this framework, and the role of off-target effects of antipsychotics, particularly at serotonergic, histaminergic, cholinergic, and adrenergic receptors for efficacy and side-effects such as weight gain, sedation and dysphoria. We review the neurobiology of schizophrenia relevant to the mode of action of antipsychotics, and for the identification of new treatment targets. This shows elevated striatal dopamine synthesis and release capacity in dorsal regions of the striatum underlies the positive symptoms of psychosis and suggests reduced dopamine release in cortical regions contributes to cognitive and negative symptoms. Current drugs act downstream of the major dopamine abnormalities in schizophrenia, and potentially worsen cortical dopamine function. We consider new approaches including targeting dopamine synthesis and storage, autoreceptors, and trace amine receptors, and the cannabinoid, muscarinic, GABAergic and glutamatergic regulation of dopamine neurons, as well as post-synaptic modulation through phosphodiesterase inhibitors. Finally, we consider treatments for cognitive and negative symptoms such dopamine agonists, nicotinic agents and AMPA modulators before discussing immunological approaches which may be disease modifying. This article is part of the issue entitled 'Special Issue on Antipsychotics'.
Collapse
Affiliation(s)
- Stephen J Kaar
- Department of Psychosis Studies, 5th Floor, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, PO63 De Crespigny Park, London, SE5 8AF, United Kingdom.
| | - Sridhar Natesan
- Department of Psychosis Studies, 5th Floor, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, PO63 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Robert McCutcheon
- Department of Psychosis Studies, 5th Floor, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, PO63 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Oliver D Howes
- Department of Psychosis Studies, 5th Floor, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, PO63 De Crespigny Park, London, SE5 8AF, United Kingdom.
| |
Collapse
|
27
|
Effect of CYP2D6 genotype on exposure and efficacy of risperidone and aripiprazole: a retrospective, cohort study. Lancet Psychiatry 2019; 6:418-426. [PMID: 31000417 DOI: 10.1016/s2215-0366(19)30088-4] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND The polymorphic CYP2D6 enzyme metabolises the antipsychotic drugs risperidone and aripiprazole to their active metabolites, 9OH-risperidone and dehydroaripiprazole. The aim of this study was to quantify the effect of CYP2D6 genetic variability on risperidone and aripiprazole exposure and treatment in a large patient population. METHODS We retrospectively obtained patient data from a routine therapeutic drug monitoring database at the Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway, between Jan 1, 2005, and Oct 15, 2018. Individuals included in our analyses were CYP2D6-genotyped patients treated with risperidone or aripiprazole. Inclusion criteria for measurement of pharmacokinetic parameters (drug and metabolite serum concentrations) were oral administration of risperidone or aripiprazole, information known about prescribed daily dose and comedications, and aged older than 18 years. Exclusion criteria included polypharmacy with drugs known to be CYP2D6 inhibitors or CYP3A4 inducers or inhibitors. Treatment failure was analysed in all patients treated with risperidone or aripiprazole without these criteria. The first endpoint in our analysis was the metabolism of risperidone to 9OH-risperidone and aripiprazole to dehydroaripiprazole, estimated by the log-transformed ratio between the concentrations of metabolite and parent drug (ie, the metabolic ratio for risperidone [9OH-risperidone]/[risperidone] and the metabolic ratio for aripiprazole [dehydroaripiprazole]/[aripiprazole]). Endpoint two was measurement of drug exposure, quantified by the dose-normalised sum of parent drug and active metabolite serum concentrations (ie, active moiety). The third endpoint of treatment failure was measured as the number of patients switched from risperidone or aripiprazole to another antipsychotic drug within 1 year after the last therapeutic drug monitoring analysis of risperidone or aripiprazole. Patient subgroups were defined by CYP2D6 genotype-determined metaboliser status: poor metabolisers, intermediate metabolisers, normal metabolisers, and ultrarapid metabolisers. ANOVA was used to assess the differences in metabolic ratios, active moieties, and daily doses between individual metaboliser categories, and risperidone and aripiprazole therapeutic failures were compared by logistic regression using the normal metaboliser subgroup as a reference. FINDINGS 1288 risperidone-treated patients and 1334 aripiprazole-treated patients were included in the study, of whom 725 (56%) risperidone-treated and 890 (67%) aripiprazole-treated patients were eligible for the pharmacokinetic analyses. CYP2D6 genotype significantly changed risperidone and aripiprazole metabolism resulting in an approximately 1·6-times and 1·4-times increase in risperidone and aripiprazole active moiety exposure in poor and intermediate metabolisers compared with normal metabolisers, respectively (odds ratios [OR] for the risperidone dose-normalised active moiety concentration 1·568, 95% CI 1·401-1·736, and 1·373, 1·213-1·532; and for the aripiprazole dose-normalised active moiety concentration 1·585, 1·447-1·724, and 1·476, 1·263-1·688, respectively; p<0·0001 for all). Compared with doses for normal metabolisers, clinicians reduced daily doses of risperidone and aripiprazole administered to poor metabolisers by 19% (95% CI 5-35, p=0·010) and 15% (95% CI 1-28, p=0·033) respectively. The incidence of switching from risperidone to another antipsychotic was increased in ultrarapid metabolisers (OR 2·934, 95% CI 1·437-5·989, p=0·003) and poor metabolisers (1·874, 1·128-3·112, p=0·015); by contrast, the incidence of switching from aripiprazole to another antipsychotic was not significantly related to CYP2D6 metaboliser status. INTERPRETATION CYP2D6 genotype had a substantial clinical effect on risperidone and aripiprazole exposure and on the therapeutic failure of risperidone. Pre-emptive CYP2D6 genotyping would be valuable for individualising risperidone and aripiprazole dosing and treatment optimisation. FUNDING H2020 program U-PGx, The Swedish Research Council, the Swedish Brain foundation, and the South-Eastern Norway Regional Health Authority.
Collapse
|
28
|
Aguilar L, Lorenzo C, Fernández-Ovejero R, Roncero C, Montejo AL. Tardive Dyskinesia After Aripiprazole Treatment That Improved With Tetrabenazine, Clozapine, and Botulinum Toxin. Front Pharmacol 2019; 10:281. [PMID: 30949057 PMCID: PMC6435569 DOI: 10.3389/fphar.2019.00281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/06/2019] [Indexed: 11/13/2022] Open
Abstract
We report on a patient with tardive dyskinesia (TDK) treated with aripiprazole, a third-generation antipsychotic with partial D2 agonist-antagonist activity at both the dopamine and serotonin receptors. The patient’s condition improved with administration of a combination of tetrabenazine, botulinum toxin, and clozapine, which has previously not been used. We suggest that this treatment combination may have potential benefits for patients with TDK. After aripiprazole discontinuation, the patient was treated with clozapine (150 mg/day) and biperiden (8 mg/day). Due to a lack of improvement, we administered 300 units (intramuscularly; IM) of botulinum toxin into the paravertebral muscles every 3 months and 1,000 units IM every 4 months in addition to tetrabenazine (75 mg/day) and biperiden (8 mg/day). The patient stopped this treatment, at which point TDK reappeared. After starting a treatment regimen of clozapine (100 mg/day), tetrabenazine (75 mg/day), and botulinum toxin (300 units IM), the patient’s symptoms remitted.
Collapse
Affiliation(s)
- Lourdes Aguilar
- Psychiatry Service, Health Care Complex, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Psychiatry, University of Salamanca, Salamanca, Spain
| | - Carolina Lorenzo
- Psychiatry Service, Health Care Complex, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Psychiatry, University of Salamanca, Salamanca, Spain
| | | | - Carlos Roncero
- Psychiatry Service, Health Care Complex, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Psychiatry, University of Salamanca, Salamanca, Spain
| | - Angel L Montejo
- Psychiatry Service, Health Care Complex, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Nursing School E.U.E.F., University of Salamanca, Salamanca, Spain
| |
Collapse
|
29
|
Dias KCF, de Almeida JC, Vasconcelos LC, Patrocínio MLV, Barbosa TM, Ximenes NC, Leitão APDA, Louchard BO, Pimenta ATÁ, Pinto FDCL, Leal LKAM, Honório Junior JER, Vasconcelos SMM. Standardized extract of Erythrina velutina Willd. attenuates schizophrenia-Like behaviours and oxidative parameters in experimental animal models. J Pharm Pharmacol 2018; 71:379-389. [PMID: 30456833 DOI: 10.1111/jphp.13039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/19/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To study the effects of the standardized extract from the leaves of Erythrina velutina in behavioural and oxidative parameters in the ketamine-induced schizophrenia model. METHODS Mice received ketamine (KET) or saline for 7 days. From 8th to 14th day, the animals received Erythrine (Eryt) (100, 200 or 400 mg/kg) or olanzapine (Olanz), 1 h after KET administration. At 14th day, 30 min after the last administration of KET, the open-field and pre-pulse inhibition (PPI) tests were performed. Then, the animals were sacrificed and the prefrontal cortex (PFC), hippocampus (HC) and striatum (ST) were dissected for the oxidative tests. KEY FINDINGS Ketamine increased spontaneous locomotor activity and grooming. KET decreased the PPI, which was reversed by combining it with Eryt or olanzapine. KET decreased GSH concentration in PFC and ST this was reversed by Eryt. KET increased MDA concentration in PFC and HC this was reversed by Eryt. Eryt and Olanzapine reduced MDA concentration in ST when compared to KET group. Nitrite concentration was reduced by administration of KET in the PFC. CONCLUSIONS These results demonstrate that the standardized extract of E. velutina can prevent behavioural symptoms and oxidative stress induced by repeated doses of KET.
Collapse
Affiliation(s)
- Katia Cilene Ferreira Dias
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil
| | - Jamily Cunha de Almeida
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil
| | - Luna Costa Vasconcelos
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil
| | | | - Talita Matias Barbosa
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil
| | - Naiara Coelho Ximenes
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil
| | | | | | | | | | | | - José Eduardo Ribeiro Honório Junior
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Ceará, Brazil.,School of Medicine, University Center Christus-Unichristus, Ceará, Brazil
| | | |
Collapse
|
30
|
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
|
31
|
Shin S, Kim S, Seo S, Lee JS, Howes OD, Kim E, Kwon JS. The relationship between dopamine receptor blockade and cognitive performance in schizophrenia: a [ 11C]-raclopride PET study with aripiprazole. Transl Psychiatry 2018; 8:87. [PMID: 29686254 PMCID: PMC5913226 DOI: 10.1038/s41398-018-0134-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 02/13/2018] [Accepted: 02/22/2018] [Indexed: 12/02/2022] Open
Abstract
Aripiprazole's effects on cognitive function in patients with schizophrenia are unclear because of the difficulty in disentangling specific effects on cognitive function from secondary effects due to the improvement in other schizophrenic symptoms. One approach to address this is to use an intermediate biomarker to investigate the relationship between the drug's effect on the brain and change in cognitive function. This study aims to investigate aripiprazole's effect on working memory by determining the correlation between dopamine D2/3 (D2/3) receptor occupancy and working memory of patients with schizophrenia. Seven patients with schizophrenia participated in the study. Serial positron emission tomography (PET) scans with [11C]raclopride were conducted at 2, 26, and 74 h after the administration of aripiprazole. The subjects performed the N-back task just after finishing the [11C]raclopride PET scan. The mean (±SD) D2/3 receptor occupancies were 66.9 ± 6.7% at 2 h, 65.0 ± 8.6% at 26, and 57.7 ± 11.2% at 74 h after administering aripiprazole. Compared with performance on the zero-back condition, performance in memory-loaded conditions (one-, two-, and three-back conditions) was significantly related to D2/3 receptor occupancy by aripiprazole (error rate: ß = -2.236, t = -6.631, df = 53.947, and p = 0.001; reaction time: ß = -9.567, t = -2.808, df = 29.967, and p = 0.009). Although the sample size was relatively small, these results suggest that aripiprazole as a dopamine-partial agonist could improve cognitive function in patients with schizophrenia.
Collapse
Affiliation(s)
- Sangho Shin
- 0000 0004 0647 3378grid.412480.bDepartment of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggi-do, 13620 Republic of Korea
| | - Seoyoung Kim
- 0000 0004 0647 3378grid.412480.bDepartment of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggi-do, 13620 Republic of Korea
| | - Seongho Seo
- 0000 0004 0470 5905grid.31501.36Department of Brain and Cognitive Sciences, College of Natural Science, Seoul National University, Seoul, 08826 Republic of Korea ,0000 0004 0470 5905grid.31501.36Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Jae Sung Lee
- 0000 0004 0470 5905grid.31501.36Department of Brain and Cognitive Sciences, College of Natural Science, Seoul National University, Seoul, 08826 Republic of Korea ,0000 0004 0470 5905grid.31501.36Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Oliver D. Howes
- 0000 0001 2322 6764grid.13097.3cInstitute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF UK ,0000000122478951grid.14105.31Medical Research Council Clinical Sciences Centre, London, W12 0NN UK ,0000 0001 0705 4923grid.413629.bImperial College London, Hammersmith Hospital Campus, London, W12 0NN UK
| | - Euitae Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggi-do, 13620, Republic of Korea. .,Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Jun Soo Kwon
- 0000 0004 0470 5905grid.31501.36Department of Brain and Cognitive Sciences, College of Natural Science, Seoul National University, Seoul, 08826 Republic of Korea ,0000 0004 0470 5905grid.31501.36Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea ,0000 0001 0302 820Xgrid.412484.fDepartment of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080 Republic of Korea ,0000 0004 0470 5905grid.31501.36Institute of Human Behavioral Medicine, SNU-MRC, Seoul, 03080 Republic of Korea
| |
Collapse
|
32
|
Plitman E, Chavez S, Nakajima S, Iwata Y, Chung JK, Caravaggio F, Kim J, Alshehri Y, Chakravarty MM, De Luca V, Remington G, Gerretsen P, Graff-Guerrero A. Striatal neurometabolite levels in patients with schizophrenia undergoing long-term antipsychotic treatment: A proton magnetic resonance spectroscopy and reliability study. Psychiatry Res Neuroimaging 2018; 273:16-24. [PMID: 29414127 DOI: 10.1016/j.pscychresns.2018.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/14/2017] [Accepted: 01/22/2018] [Indexed: 12/13/2022]
Abstract
Previous proton magnetic resonance spectroscopy (1H-MRS) studies have reported disrupted levels of various neurometabolites in patients with schizophrenia. An area of particular interest within this patient population is the striatum, which is highly implicated in the pathophysiology of schizophrenia. The present study examined neurometabolite levels in the striatum of 12 patients with schizophrenia receiving antipsychotic treatment for at least 1 year and 11 healthy controls using 3-Tesla 1H-MRS (PRESS, TE = 35 ms). Glutamate, glutamate+glutamine (Glx), myo-inositol, choline, N-acetylaspartate, and creatine levels were estimated using LCModel, and corrected for fraction of cerebrospinal fluid in the 1H-MRS voxel. Striatal neurometabolite levels were compared between groups. Multiple study visits permitted a reliability assessment for neurometabolite levels (days between paired 1H-MRS acquisitions: average = 90.33; range = 7-306). Striatal neurometabolite levels did not differ between groups. Within the whole sample, intraclass correlation coefficients for glutamate, Glx, myo-inositol, choline, and N-acetylaspartate were fair to excellent (0.576-0.847). The similarity in striatal neurometabolite levels between groups implies a marked difference from the antipsychotic-naïve first-episode state, especially in terms of glutamatergic neurometabolites, and might provide insight regarding illness progression and the influence of antipsychotic medication.
Collapse
Affiliation(s)
- Eric Plitman
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Sofia Chavez
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Shinichiro Nakajima
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Neuropsychiatry, Keio University, Tokyo, Japan
| | - Yusuke Iwata
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jun Ku Chung
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Julia Kim
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Youssef Alshehri
- Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Departments of Psychiatry and Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Vincenzo De Luca
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Schizophrenia Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Institute Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Gary Remington
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Schizophrenia Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Institute Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Institute Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Abstract
Dopamine D3 receptors have key roles in behavioral reward, addiction, Parkinson's disease, and schizophrenia, and there is interest in studying their role in these disorders using PET. However, current PET radiotracers for studying D3 receptors in humans all bind to both D2 and D3 due to similarities between the two receptors. Selective D2 and D3 radioligands would aid investigation of the differences between D2 and D3 circuitry in the central nervous system. While there are currently in vitro measures of ligand D3/D2 selectivity, there is a need for an in vivo PET measure of D3/D2 selectivity. This review discusses current PET imaging of dopamine D2/D3 receptors and proposes methodology for quantitating in vivo selectivity of probes for PET imaging of dopamine D3 receptors.
Collapse
Affiliation(s)
- Robert K Doot
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyle J Labban
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
34
|
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
|
35
|
Reeves S, Eggleston K, Cort E, McLachlan E, Brownings S, Nair A, Greaves S, Smith A, Dunn J, Marsden P, Kessler R, Taylor D, Bertrand J, Howard R. Therapeutic D2/3 receptor occupancies and response with low amisulpride blood concentrations in very late-onset schizophrenia-like psychosis (VLOSLP). Int J Geriatr Psychiatry 2018. [PMID: 28643852 DOI: 10.1002/gps.4758] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Antipsychotic drug sensitivity in very late-onset schizophrenia-like psychosis (VLOSLP) is well documented, but poorly understood. This study aimed to investigate blood drug concentration, D2/3 receptor occupancy and outcome in VLOSLP during open amisulpride prescribing, and compare this with Alzheimer's disease (AD). METHODS Blood drug concentration, prolactin, symptoms and extrapyramidal side-effects (EPS) were serially assessed during dose titration. [18 F]fallypride imaging was used to quantify D2/3 receptor occupancy. Average steady-state amisulpride concentration (Caverage, ng/ml) was estimated by incorporating pharmacokinetic (PK) data into an existing population PK model (25 AD participants, 20 healthy older people). RESULTS Eight patients (target 20) were recruited (six women; 76 + - 6 years; six treatment compliant; five serially sampled; three with paired imaging data). Mean + - SD symptom reduction was 74 ± 12% (50-100 mg/day; 92.5 + -39.4 ng/ml). Mild EPS emerged at 96 ng/ml (in AD, severe EPS, 50 mg/day, 60 ng/ml). In three participants, imaged during optimal treatment (50 mg/day; 41-70 ng/ml), caudate occupancy was 44-59% (58-74% in AD across a comparable Caverage). CONCLUSIONS Despite the small sample size, our findings are highly relevant as they suggest that, as in AD, 50 mg/day amisulpride is associated with >40% occupancy and clinically relevant responses in VLOSLP. It was not possible to fully characterise concentration-occupancy relationships in VLOSLP, and it is thus unclear whether the greater susceptibility of those with AD to emergent EPS was accounted for by increased central drug access. Further investigation of age- and diagnosis-specific threshold sensitivities is warranted, to guide amisulpride prescribing in older people, and therapeutic drug monitoring studies offer a potentially informative future approach. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Suzanne Reeves
- Division of Psychiatry, University College London, UK.,Department of Old Age Psychiatry, Kings College London, UK
| | - Kate Eggleston
- Department of Old Age Psychiatry, Kings College London, UK
| | - Elizabeth Cort
- Department of Old Age Psychiatry, Kings College London, UK
| | - Emma McLachlan
- Department of Old Age Psychiatry, Kings College London, UK
| | | | - Akshay Nair
- Division of Psychiatry, University College London, UK
| | - Suki Greaves
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Alan Smith
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Joel Dunn
- Division of Imaging Sciences, St Thomas Hospital, London, UK
| | - Paul Marsden
- Division of Imaging Sciences, St Thomas Hospital, London, UK
| | | | - David Taylor
- Division of Imaging Sciences, St Thomas Hospital, London, UK
| | - Julie Bertrand
- UMR 1137 IAME INSERM University Paris 7, France and Genetics Institute, University College London, UK
| | - Robert Howard
- Division of Psychiatry, University College London, UK.,Department of Old Age Psychiatry, Kings College London, UK
| |
Collapse
|
36
|
Reeves S, McLachlan E, Bertrand J, D'Antonio F, Brownings S, Nair A, Greaves S, Smith A, Taylor D, Dunn J, Marsden P, Kessler R, Howard R. Therapeutic window of dopamine D2/3 receptor occupancy to treat psychosis in Alzheimer's disease. Brain 2017; 140:1117-1127. [PMID: 28334978 DOI: 10.1093/brain/aww359] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022] Open
Abstract
See Caravaggio and Graff-Guerrero (doi:10.1093/awx023) for a scientific commentary on this article.Antipsychotic drugs, originally developed to treat schizophrenia, are used to treat psychosis, agitation and aggression in Alzheimer's disease. In the absence of dopamine D2/3 receptor occupancy data to inform antipsychotic prescribing for psychosis in Alzheimer's disease, the mechanisms underpinning antipsychotic efficacy and side effects are poorly understood. This study used a population approach to investigate the relationship between amisulpride blood concentration and central D2/3 occupancy in older people with Alzheimer's disease by combining: (i) pharmacokinetic data (280 venous samples) from a phase I single (50 mg) dose study in healthy older people (n = 20, 65-79 years); (ii) pharmacokinetic, 18F-fallypride D2/3 receptor imaging and clinical outcome data on patients with Alzheimer's disease who were prescribed amisulpride (25-75 mg daily) to treat psychosis as part of an open study (n = 28; 69-92 years; 41 blood samples, five pretreatment scans, 19 post-treatment scans); and (iii) 18F-fallypride imaging of an antipsychotic free Alzheimer's disease control group (n = 10, 78-92 years), to provide additional pretreatment data. Non-linear mixed effects modelling was used to describe pharmacokinetic-occupancy curves in caudate, putamen and thalamus. Model outputs were used to estimate threshold steady state blood concentration and occupancy required to elicit a clinically relevant response (>25% reduction in scores on delusions, hallucinations and agitation domains of the Neuropsychiatric Inventory) and extrapyramidal side effects (Simpson Angus Scale scores > 3). Average steady state blood levels were low (71 ± 30 ng/ml), and associated with high D2/3 occupancies (65 ± 8%, caudate; 67 ± 11%, thalamus; 52 ± 11%, putamen). Antipsychotic clinical response occurred at a threshold concentration of 20 ng/ml and D2/3 occupancies of 43% (caudate), 25% (putamen), 43% (thalamus). Extrapyramidal side effects (n = 7) emerged at a threshold concentration of 60 ng/ml, and D2/3 occupancies of 61% (caudate), 49% (putamen) and 69% (thalamus). This study has established that, as in schizophrenia, there is a therapeutic window of D2/3 receptor occupancy for optimal treatment of psychosis in Alzheimer's disease. We have also shown that occupancies within and beyond this window are achieved at very low amisulpride doses in Alzheimer's disease due to higher than anticipated occupancies for a given blood drug concentration. Our findings support a central pharmacokinetic contribution to antipsychotic sensitivity in Alzheimer's disease and implicate the blood-brain barrier, which controls central drug access. Whether high D2/3 receptor occupancies are primarily accounted for by age- or disease-specific blood-brain barrier disruption is unclear, and this is an important future area of future investigation, as it has implications beyond antipsychotic prescribing.
Collapse
Affiliation(s)
- Suzanne Reeves
- Division of Psychiatry, 149 Tottenham Court Road, London W1T 7NF, University College London, UK.,Department of Old Age Psychiatry, London, SE58AF, Kings College London, UK
| | - Emma McLachlan
- Department of Old Age Psychiatry, London, SE58AF, Kings College London, UK
| | - Julie Bertrand
- UMR 1137 IAME INSERM University Paris 7, France; and Genetics Institute, WC1E6BT, University College London, UK
| | - Fabrizia D'Antonio
- Division of Psychiatry, 149 Tottenham Court Road, London W1T 7NF, University College London, UK.,Department of Old Age Psychiatry, London, SE58AF, Kings College London, UK
| | - Stuart Brownings
- Division of Psychiatry, 149 Tottenham Court Road, London W1T 7NF, University College London, UK
| | - Akshay Nair
- Division of Psychiatry, 149 Tottenham Court Road, London W1T 7NF, University College London, UK
| | - Suki Greaves
- South London and Maudsley NHS Foundation Trust, London, SE58AZ, UK
| | - Alan Smith
- South London and Maudsley NHS Foundation Trust, London, SE58AZ, UK
| | - David Taylor
- South London and Maudsley NHS Foundation Trust, London, SE58AZ, UK
| | - Joel Dunn
- Division of Imaging Sciences, St Thomas Hospital, London, SE17EH, UK
| | - Paul Marsden
- Division of Imaging Sciences, St Thomas Hospital, London, SE17EH, UK
| | | | - Robert Howard
- Division of Psychiatry, 149 Tottenham Court Road, London W1T 7NF, University College London, UK.,Department of Old Age Psychiatry, London, SE58AF, Kings College London, UK
| |
Collapse
|
37
|
Eisenstein SA, Bogdan R, Chen L, Moerlein SM, Black KJ, Perlmutter JS, Hershey T, Barch DM. Preliminary evidence that negative symptom severity relates to multilocus genetic profile for dopamine signaling capacity and D2 receptor binding in healthy controls and in schizophrenia. J Psychiatr Res 2017; 86:9-17. [PMID: 27886638 PMCID: PMC5272837 DOI: 10.1016/j.jpsychires.2016.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/07/2016] [Accepted: 11/15/2016] [Indexed: 12/31/2022]
Abstract
Deficits in central, subcortical dopamine (DA) signaling may underlie negative symptom severity, particularly anhedonia, in healthy individuals and in schizophrenia. To investigate these relationships, we assessed negative symptoms with the Schedule for the Assessment of Negative Symptoms and the Brief Negative Symptom Scale (BNSS) and self-reported anhedonia with the Scales for Physical and Social Anhedonia (SPSA), Temporal Experience of Pleasure Scale, and Snaith-Hamilton Pleasure Scale in 36 healthy controls (HC), 27 siblings (SIB) of individuals with schizophrenia, and 66 individuals with schizophrenia or schizoaffective disorder (SCZ). A subset of participants (N = 124) were genotyped for DA-related polymorphisms in genes for DRD4, DRD2/ANKK1, DAT1, and COMT, which were used to construct biologically-informed multi-locus genetic profile (MGP) scores reflective of subcortical dopaminergic signaling. DA receptor type 2 (D2R) binding was assessed among a second subset of participants (N = 23) using PET scans with the D2R-selective, non-displaceable radioligand (N-[11C]methyl)benperidol. Higher MGP scores, reflecting elevated subcortical dopaminergic signaling capacity, were associated with less negative symptom severity, as measured by the BNSS, across all participants. In addition, higher striatal D2R binding was associated with less physical and social anhedonia, as measured by the SPSA, across HC, SIB, and SCZ. The current preliminary findings support the hypothesis that subcortical DA function may contribute to negative symptom severity and self-reported anhedonia, independent of diagnostic status.
Collapse
Affiliation(s)
- Sarah A. Eisenstein
- Psychiatry Department, Washington University School of Medicine, St. Louis, MO, USA,Radiology Department, Washington University School of Medicine, St. Louis, MO, USA,Corresponding author, Sarah A. Eisenstein, Psychiatry Department, Campus Box 8225, Washington University School of Medicine, St. Louis, MO 63110, Phone: (314) 362-7107, Fax: (314) 362-0168,
| | - Ryan Bogdan
- Psychological & Brain Sciences Department, Washington University in St. Louis, St. Louis, MO, USA.
| | - Ling Chen
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA.
| | - Stephen M. Moerlein
- Radiology Department, Washington University School of Medicine, St. Louis, MO, USA,Biochemistry Department, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin J. Black
- Psychiatry Department, Washington University School of Medicine, St. Louis, MO, USA,Radiology Department, Washington University School of Medicine, St. Louis, MO, USA,Neurology Department, Washington University School of Medicine, St. Louis, MO, USA,Neuroscience Department, Washington University School of Medicine, MO, USA
| | - Joel S. Perlmutter
- Radiology Department, Washington University School of Medicine, St. Louis, MO, USA,Biochemistry Department, Washington University School of Medicine, St. Louis, MO, USA,Programs in Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Tamara Hershey
- Psychiatry Department, Washington University School of Medicine, St. Louis, MO, USA; Radiology Department, Washington University School of Medicine, St. Louis, MO, USA; Psychological & Brain Sciences Department, Washington University in St. Louis, St. Louis, MO, USA; Neurology Department, Washington University School of Medicine, St. Louis, MO, USA.
| | - Deanna M. Barch
- Psychiatry Department, Washington University School of Medicine, St. Louis, MO, USA,Radiology Department, Washington University School of Medicine, St. Louis, MO, USA,Psychological & Brain Sciences Department, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
38
|
Topolov MK, Getova DP. Cognitive Impairment in Schizophrenia, Neurotransmitters and the New Atypical Antipsychotic Aripiprazole. Folia Med (Plovdiv) 2017; 58:12-8. [PMID: 27383873 DOI: 10.1515/folmed-2016-0002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 02/04/2016] [Indexed: 11/15/2022] Open
Abstract
Cognition is a group of mental processes that includes the capacity to perceive, think, learn and to study, and the capacity of the brain to analyze information and program adaptive behaviour. Although there has been an appreciable evolution in the therapy of psychoses in the last twenty-five years, cognitive disturbances still persist in spite of antipsychotic treatment. The cognitive decay disrupts the ability of clinically diagnosed patients with psychoses, mainly schizophrenia, to learn and to memorize skills that are useful for their family and social relationships. Moreover, cognitive deficiency is often considered to be crucial for further rehabilitation. In atypical antipsychotics there are big differences in the effects on cognitive functions. Some clinical studies demonstrate the benefits of a third generation of antipsychotics on cognitive functions in patients treated for mental illnesses. In the present study we have reviewed many articles investigating the influence of aripiprazole on cognition in human and animal subjects. Aripiprazole is a third generation antipsychotic drug that possesses a unique pharmacodynamic profile, which in conjunction with recently published scientific data on the drugs' influence on antidepressant, anxiolytic and cognitive functions, suggests a highly positive future potential for restorative cognitive treatment and ongoing healthy function. The data included in the review will contribute to determining the potential benefits of aripiprazole on memory and training processes.
Collapse
Affiliation(s)
- Mariyan K Topolov
- Department of Pharmacology and Drug Toxicology, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Damianka P Getova
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
39
|
Weinstein JJ, Chohan MO, Slifstein M, Kegeles LS, Moore H, Abi-Dargham A. Pathway-Specific Dopamine Abnormalities in Schizophrenia. Biol Psychiatry 2017; 81:31-42. [PMID: 27206569 PMCID: PMC5177794 DOI: 10.1016/j.biopsych.2016.03.2104] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 03/21/2016] [Accepted: 03/25/2016] [Indexed: 02/08/2023]
Abstract
In light of the clinical evidence implicating dopamine in schizophrenia and the prominent hypotheses put forth regarding alterations in dopaminergic transmission in this disease, molecular imaging has been used to examine multiple aspects of the dopaminergic system. We review the imaging methods used and compare the findings across the different molecular targets. Findings have converged to suggest early dysregulation in the striatum, especially in the rostral caudate, manifesting as excess synthesis and release. Recent data showed deficit extending to most cortical regions and even to other extrastriatal subcortical regions not previously considered to be "hypodopaminergic" in schizophrenia. These findings yield a new topography for the dopaminergic dysregulation in schizophrenia. We discuss the dopaminergic innervation within the individual projection fields to provide a topographical map of this dual dysregulation and explore potential cellular and circuit-based mechanisms for brain region-dependent alterations in dopaminergic parameters. This refined knowledge is essential to better guide translational studies and efforts in early drug development.
Collapse
Affiliation(s)
- Jodi J. Weinstein
- Columbia University Department of Psychiatry, New York, NY,New York State Psychiatric Institute Division of Translational Imaging,Corresponding author: Jodi Weinstein, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 31, New York, New York 10032, +1-646-774-8123,
| | - Muhammad O. Chohan
- New York State Psychiatric Institute Division of Integrative Neuroscience
| | - Mark Slifstein
- Columbia University Department of Psychiatry, New York, NY,New York State Psychiatric Institute Division of Translational Imaging
| | - Lawrence S. Kegeles
- Columbia University Department of Psychiatry, New York, NY,New York State Psychiatric Institute Division of Translational Imaging
| | - Holly Moore
- Columbia University Department of Psychiatry, New York, NY,New York State Psychiatric Institute Division of Integrative Neuroscience
| | - Anissa Abi-Dargham
- Columbia University Department of Psychiatry, New York, NY,New York State Psychiatric Institute Division of Translational Imaging
| |
Collapse
|
40
|
Holmes SE, Hinz R, Drake RJ, Gregory CJ, Conen S, Matthews JC, Anton-Rodriguez JM, Gerhard A, Talbot PS. In vivo imaging of brain microglial activity in antipsychotic-free and medicated schizophrenia: a [ 11C](R)-PK11195 positron emission tomography study. Mol Psychiatry 2016; 21:1672-1679. [PMID: 27698434 DOI: 10.1038/mp.2016.180] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) imaging of the 18 kDa translocator protein (TSPO) has been used to investigate whether microglial activation, an indication of neuroinflammation, is evident in the brain of adults with schizophrenia. Interpretation of these studies is confounded by potential modulatory effects of antipsychotic medication on microglial activity. In the first such study in antipsychotic-free schizophrenia, we have used [11C](R)-PK11195 PET to compare TSPO availability in a predominantly antipsychotic-naive group of moderate-to-severely symptomatic unmedicated patients (n=8), similarly symptomatic medicated patients with schizophrenia taking risperidone or paliperidone by regular intramuscular injection (n=8), and healthy comparison subjects (n=16). We found no evidence for increased TSPO availability in antipsychotic-free patients compared with healthy controls (mean difference 4%, P=0.981). However, TSPO availability was significantly elevated in medicated patients (mean increase 88%, P=0.032) across prefrontal (dorsolateral, ventrolateral, orbital), anterior cingulate and parietal cortical regions. In the patients, TSPO availability was also strongly correlated with negative symptoms measured using the Positive and Negative Syndrome Scale across all the brain regions investigated (r=0.651-0.741). We conclude that the pathophysiology of schizophrenia is not associated with microglial activation in the 2-6 year period following diagnosis. The elevation in the medicated patients may be a direct effect of the antipsychotic, although this study cannot exclude treatment resistance and/or longer illness duration as potential explanations. It also remains to be determined whether it is present only in a subset of patients, represents a pro- or anti-inflammatory state, its association with primary negative symptoms, and whether there are significant differences between antipsychotics.
Collapse
Affiliation(s)
- S E Holmes
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - R Hinz
- Wolfson Molecular Imaging Centre, Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - R J Drake
- Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - C J Gregory
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - S Conen
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - J C Matthews
- Wolfson Molecular Imaging Centre, Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - J M Anton-Rodriguez
- Wolfson Molecular Imaging Centre, Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Gerhard
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - P S Talbot
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
41
|
Abstract
This safety assessment provides a detailed analysis of key studies and focuses on the six most widely used antipsychotic drugs. Lines of evidence include mechanisms of action, short-term treatment of psychosis, relapse prevention, early intervention in schizophrenia, long-term comparisons between first- and second-generation agents, and flexible treatment algorithms. Despite the diversity of study settings, several common features were seen. All the agents obstruct normal signaling through widely dispersed dopamine D2 receptors. Treatment failure or psychosis relapse was the most frequent outcome in most key studies, ranging from 38 to 93%. High discontinuation rates caused most trials to fail to demonstrate a substantial treatment benefit, or difference from an active comparator. Assessment of harm to the extrapyramidal motor system was confounded because of extensive neurological impairment from previous antipsychotic drug treatment measured at baseline, abrupt discontinuation effects, and high rates of concomitant medications to manage drug adverse effects. Claims that second-generation antipsychotic drugs have safety advantages over classical neuroleptic drugs and prevent relapse were not supported in these key studies. The extent of injury to and impairment of multiple body systems caused by antipsychotic drugs shows the need for a scientific, clinical, and regulatory reappraisal of the appropriate use of these agents.
Collapse
Affiliation(s)
- Thomas J Moore
- Institute for Safe Medication Practices, 101 N. Columbus St, Suite 410, Alexandria, VA, 22314, USA. .,Department of Epidemiology and Biostatistics, George Washington University Milken Institute School of Public Health, Washington, DC, USA.
| | - Curt D Furberg
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
42
|
A Case of Aripiprazole-Induced Tardive Dyskinesia with Dramatic Evolution. Case Rep Psychiatry 2016; 2016:7031245. [PMID: 27818825 PMCID: PMC5080510 DOI: 10.1155/2016/7031245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/23/2016] [Accepted: 09/26/2016] [Indexed: 11/17/2022] Open
Abstract
Aripiprazole is reported to be a good clinical safety profile antipsychotic. However, recent data suggest that the risk of tardive dyskinesia could be higher than initially thought. We report the case of aripiprazole-induced tardive dyskinesia with dramatic evolution in a patient with several risk factors, including older age and exposure to antipsychotic over a period longer than six months. This case and its dramatic evolution, associated with other cases recently published, suggest reconsidering the real risk of tardive dyskinesia associated with aripiprazole, particularly in the elderly.
Collapse
|
43
|
Gomeni R, Bressolle-Gomeni F, Fava M. Response Surface Analysis and Nonlinear Optimization Algorithm for Maximization of Clinical Drug Performance: Application to Extended-Release and Long-Acting Injectable Paliperidone. J Clin Pharmacol 2016; 56:1296-306. [PMID: 26899406 DOI: 10.1002/jcph.724] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/15/2016] [Indexed: 11/07/2022]
Abstract
Model-based approach is recognized as a tool to make drug development more productive and to better support regulatory and therapeutic decisions. The objective of this study was to develop a novel model-based methodology based on the response surface analysis and a nonlinear optimizer algorithm to maximize the clinical performances of drug treatments. The treatment response was described using a drug-disease model accounting for multiple components such as the dosage regimen, the pharmacokinetic characteristics of a drug (including the mechanism and the rate of drug delivery), and the exposure-response relationship. Then, the clinical benefit of a treatment was defined as a function of the diseases and the clinical endpoints and was estimated as a function of the target pharmacodynamic endpoints used to evaluate the treatment effect. A case study is presented to illustrate how the treatment performances of paliperidone extended release (ER) and paliperidone long-acting injectable (LAI) can be improved. A convolution-based approach was used to characterize the pharmacokinetics of ER and LAI paliperidone. The drug delivery properties and the dosage regimen maximizing the clinical benefit (defined as the target level of D2 receptor occupancy) were estimated using a nonlinear optimizer. The results of the analysis indicated that a substantial improvement in clinical benefit (from 15% to 27% for the optimization of the in vivo release and from ∼30% to ∼70% for the optimization of dosage regimen) was obtained when optimal strategies were deployed either for optimizing the in vivo drug delivery properties of ER formulations or for optimizing the dosage regimen of LAI formulations.
Collapse
Affiliation(s)
- Roberto Gomeni
- R&D Department, Pharmacometrica, Longcol, La Fouillade, France.
| | | | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
44
|
Li P, Snyder GL, Vanover KE. Dopamine Targeting Drugs for the Treatment of Schizophrenia: Past, Present and Future. Curr Top Med Chem 2016; 16:3385-3403. [PMID: 27291902 PMCID: PMC5112764 DOI: 10.2174/1568026616666160608084834] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 01/18/2023]
Abstract
Schizophrenia is a chronic and debilitating neuropsychiatric disorder affecting approximately 1% of the world's population. This disease is associated with considerable morbidity placing a major financial burden on society. Antipsychotics have been the mainstay of the pharmacological treatment of schizophrenia for decades. The traditional typical and atypical antipsychotics demonstrate clinical efficacy in treating positive symptoms, such as hallucinations and delusions, while are largely ineffective and may worsen negative symptoms, such as blunted affect and social withdrawal, as well as cognitive function. The inability to treat these latter symptoms may contribute to social function impairment associated with schizophrenia. The dysfunction of multiple neurotransmitter systems in schizophrenia suggests that drugs selectively targeting one neurotransmission pathway are unlikely to meet all the therapeutic needs of this heterogeneous disorder. Often, however, the unintentional engagement of multiple pharmacological targets or even the excessive engagement of intended pharmacological targets can lead to undesired consequences and poor tolerability. In this article, we will review marketed typical and atypical antipsychotics and new therapeutic agents targeting dopamine receptors and other neurotransmitters for the treatment of schizophrenia. Representative typical and atypical antipsychotic drugs and new investigational drug candidates will be systematically reviewed and compared by reviewing structure-activity relationships, pharmacokinetic properties, drug metabolism and safety, pharmacological properties, preclinical data in animal models, clinical outcomes and associated side effects.
Collapse
Affiliation(s)
- Peng Li
- Intra-Cellular Therapies Inc, 430 East 29th Street, Suite 900, New York, NY 10016, United States.
| | | | | |
Collapse
|
45
|
Jarcho JM, Feier NA, Labus JS, Naliboff B, Smith SR, Hong JY, Colloca L, Tillisch K, Mandelkern MA, Mayer EA, London ED. Placebo analgesia: Self-report measures and preliminary evidence of cortical dopamine release associated with placebo response. NEUROIMAGE-CLINICAL 2015; 10:107-14. [PMID: 26759785 PMCID: PMC4683423 DOI: 10.1016/j.nicl.2015.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 11/25/2022]
Abstract
Placebo analgesia is measured by self-report, yet current, expected, and recalled efficacy may be differentially related to brain function. Here we used a human thermal pain model to compare self-reports of expected, concurrent, and recalled efficacy of a topical placebo analgesic, and tested associations of the three measures of efficacy with changes in dopamine D2/D3 receptor availability in brain using [18F]fallypride with positron emission tomography (PET). Participants (15 healthy women) were assessed on three test days. The first test day included a laboratory visit, during which the temperature needed to evoke consistent pain was determined, placebo analgesia was induced via verbal and experience-based expectation, and the placebo response was measured. On two subsequent test days, PET scans were performed in Control and Placebo conditions, respectively, in counterbalanced order. During Visit 1, concurrent and recalled placebo efficacy were unrelated; during the Placebo PET visit, expected and recalled efficacy were highly correlated (ρ = 0.68, p = 0.005), but concurrent efficacy was unrelated to expected or recalled efficacy. Region of interest analysis revealed dopamine D2/D3 receptor availability was lower in left ventrolateral prefrontal cortex in the Placebo condition (p < 0.001, uncorrected), and greater change in this measure was associated with higher levels of recalled analgesic efficacy (ρ = 0.58, p = 0.02). These preliminary findings underscore the need to consider how self-reported symptom improvement is assessed in clinical trials of analgesics and suggest that dopaminergic activity in the ventrolateral prefrontal cortex may promote recalled efficacy of placebo. Healthy women reported on expected, concurrent, and recalled placebo analgesia. Measures were obtained in the lab and during PET scanning with [18F]fallypride. Dopamine D2/D3 receptor availability in PFC was associated with placebo analgesia. This relationship was specific to recalled placebo analgesia efficacy.
Collapse
Affiliation(s)
- Johanna M Jarcho
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Psychology, Stony Brook University, NY, USA
| | - Natasha A Feier
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Alan Edwards Centre for Research on Pain, Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Jennifer S Labus
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Physiology, UCLA, Los Angeles, CA, United States; Pain and Interoception Network (PAIN), UCLA, Los Angeles, CA, Unit
| | - Bruce Naliboff
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Physiology, UCLA, Los Angeles, CA, United States; Pain and Interoception Network (PAIN), UCLA, Los Angeles, CA, Unit
| | - Suzanne R Smith
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Physiology, UCLA, Los Angeles, CA, United States; Pain and Interoception Network (PAIN), UCLA, Los Angeles, CA, Unit
| | - Jui-Yang Hong
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Physiology, UCLA, Los Angeles, CA, United States; Pain and Interoception Network (PAIN), UCLA, Los Angeles, CA, Unit
| | - Luana Colloca
- School of Nursing, University of Maryland, Baltimore, MD, USA; School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Kirsten Tillisch
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Physiology, UCLA, Los Angeles, CA, United States; Pain and Interoception Network (PAIN), UCLA, Los Angeles, CA, Unit; VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | | | - Emeran A Mayer
- Gail and Gerald Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Physiology, UCLA, Los Angeles, CA, United States; Pain and Interoception Network (PAIN), UCLA, Los Angeles, CA, Unit
| | - Edythe D London
- Department of Physiology, UCLA, Los Angeles, CA, United States; VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
46
|
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: 2.9] [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
|
47
|
Chen JX, Su YA, Bian QT, Wei LH, Zhang RZ, Liu YH, Correll C, Soares JC, Yang FD, Wang SL, Zhang XY. Adjunctive aripiprazole in the treatment of risperidone-induced hyperprolactinemia: A randomized, double-blind, placebo-controlled, dose-response study. Psychoneuroendocrinology 2015; 58:130-40. [PMID: 25981348 DOI: 10.1016/j.psyneuen.2015.04.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/27/2015] [Accepted: 04/16/2015] [Indexed: 01/28/2023]
Abstract
Hyperprolactinemia is an unwanted adverse effect associated with several antipsychotics. The addition of partial dopamine receptor agonist aripiprazole may attenuate antipsychotic-induced hyperprolactinemia effectively. However, the ideal dosing regimen for this purpose is unknown. We aimed to evaluate the dose effects of adjunctive treatment with aripiprazole on prolactin levels and hyperprolactinemia in schizophrenia patients. Stable subjects 18-45 years old with schizophrenia and hyperprolactinemia (i.e., >24 ng/ml for females and >20 ng/ml for males) were randomly assigned to receive 8 weeks of placebo (n=30) or oral aripiprazole 5mg/day (n=30), 10mg/day (n=29), or 20mg/day (n=30) added on to fixed dose risperidone treatment. Serum prolactin levels were measured at baseline and after 2, 4 and 8 weeks; clinical symptoms and side effects were assessed at baseline and week 8 using the Positive and Negative Syndrome Scale, Clinical Global Impressions Severity scale, Barnes Akathisia Scale, Simpson-Angus Scale and UKU Side Effects Rating Scale. Of 119 randomized patients, 107 (89.9%) completed the 8-week study. At study end, all three aripiprazole doses resulted in significantly lower prolactin levels (beginning at week 2), higher response rates (≥30% prolactin reduction) and higher prolactin normalization rates than placebo. Effects were significantly greater in the 10 and 20mg/day groups than the 5mg/day group. No significant changes were observed in any treatment groups regarding psychopathology and adverse effect ratings. Adjunctive aripiprazole treatment was effective and safe for resolving risperidone-induced hyperprolactinemia, producing significant and almost maximal improvements by week 2 without significant effects on psychopathology and side effects.
Collapse
Affiliation(s)
- Jing-Xu Chen
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Yun-Ai Su
- Peking University Sixth Hospital/Institute of Mental Health, Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Qing-Tao Bian
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Li-He Wei
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Rong-Zhen Zhang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Yan-Hong Liu
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Christoph Correll
- The Zucker Hillside Hospital, Psychiatry Research, North Shore-Long Island Jewish Health System, Glen Oaks, NY, USA
| | - Jair C Soares
- Department of Psychiatry and Behavioral Sciences, Harris County Psychiatric Center, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fu-De Yang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Shao-Li Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China
| | - Xiang-Yang Zhang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China; Department of Psychiatry and Behavioral Sciences, Harris County Psychiatric Center, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
48
|
Skilleter AJ, Weickert CS, Vercammen A, Lenroot R, Weickert TW. Peripheral BDNF: a candidate biomarker of healthy neural activity during learning is disrupted in schizophrenia. Psychol Med 2015; 45:841-854. [PMID: 25162472 PMCID: PMC4413857 DOI: 10.1017/s0033291714001925] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 07/16/2014] [Accepted: 07/16/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is an important regulator of synaptogenesis and synaptic plasticity underlying learning. However, a relationship between circulating BDNF levels and brain activity during learning has not been demonstrated in humans. Reduced brain BDNF levels are found in schizophrenia and functional neuroimaging studies of probabilistic association learning in schizophrenia have demonstrated reduced activity in a neural network that includes the prefrontal and parietal cortices and the caudate nucleus. We predicted that brain activity would correlate positively with peripheral BDNF levels during probabilistic association learning in healthy adults and that this relationship would be altered in schizophrenia. METHOD Twenty-five healthy adults and 17 people with schizophrenia or schizo-affective disorder performed a probabilistic association learning test during functional magnetic resonance imaging (fMRI). Plasma BDNF levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS We found a positive correlation between circulating plasma BDNF levels and brain activity in the parietal cortex in healthy adults. There was no relationship between plasma BDNF levels and task-related activity in the prefrontal, parietal or caudate regions in schizophrenia. A direct comparison of these relationships between groups revealed a significant diagnostic difference. CONCLUSIONS This is the first study to show a relationship between peripheral BDNF levels and cortical activity during learning, suggesting that plasma BDNF levels may reflect learning-related brain activity in healthy humans. The lack of relationship between plasma BDNF and task-related brain activity in patients suggests that circulating blood BDNF may not be indicative of learning-dependent brain activity in schizophrenia.
Collapse
Affiliation(s)
- A. J. Skilleter
- School of Psychiatry,
University of New South Wales, Kensington,
NSW, Australia
- Neuroscience Research Australia,
Randwick, NSW, Australia
- Schizophrenia Research Institute,
Darlinghurst, NSW, Australia
| | - C. S. Weickert
- School of Psychiatry,
University of New South Wales, Kensington,
NSW, Australia
- Neuroscience Research Australia,
Randwick, NSW, Australia
- Schizophrenia Research Institute,
Darlinghurst, NSW, Australia
| | - A. Vercammen
- School of Psychiatry,
University of New South Wales, Kensington,
NSW, Australia
- Neuroscience Research Australia,
Randwick, NSW, Australia
- Schizophrenia Research Institute,
Darlinghurst, NSW, Australia
| | - R. Lenroot
- School of Psychiatry,
University of New South Wales, Kensington,
NSW, Australia
- Neuroscience Research Australia,
Randwick, NSW, Australia
- Schizophrenia Research Institute,
Darlinghurst, NSW, Australia
| | - T. W. Weickert
- School of Psychiatry,
University of New South Wales, Kensington,
NSW, Australia
- Neuroscience Research Australia,
Randwick, NSW, Australia
- Schizophrenia Research Institute,
Darlinghurst, NSW, Australia
| |
Collapse
|
49
|
Abstract
Clinical experience with aripiprazole has confirmed the effectiveness and the safety of this novel antipsychotic drug in patients with schizophrenia as well as for the treatment of mania in type I bipolar disorder. However the generalization of the results from clinical trials requires further effort in order to address some issues and to overcome incorrect and partial interpretation of the clinical evidence. This article provides some straightforward guidance that may help clinical psychiatrists to translate the mechanism of action of aripiprazole into clinical setting, thus improving the appropriate use of the drug through rational application of its pharmacological profile. Examples of paradigmatic clinical situations are presented and discussed, suggesting possible intervention strategies, which may contribute to achieving the most appropriate use of the pharmacological properties of aripiprazole in real life settings.
Collapse
Affiliation(s)
- Guido Di Sciascio
- Department of Psychiatry, University Hospital "Policlinico", Bari, Italy
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
50
|
de Bartolomeis A, Tomasetti C, Iasevoli F. Update on the Mechanism of Action of Aripiprazole: Translational Insights into Antipsychotic Strategies Beyond Dopamine Receptor Antagonism. CNS Drugs 2015; 29:773-99. [PMID: 26346901 PMCID: PMC4602118 DOI: 10.1007/s40263-015-0278-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Dopamine partial agonism and functional selectivity have been innovative strategies in the pharmacological treatment of schizophrenia and mood disorders and have shifted the concept of dopamine modulation beyond the established approach of dopamine D2 receptor (D2R) antagonism. Despite the fact that aripiprazole was introduced in therapy more than 12 years ago, many questions are still unresolved regarding the complexity of the effects of this agent on signal transduction and intracellular pathways, in part linked to its pleiotropic receptor profile. The complexity of the mechanism of action has progressively shifted the conceptualization of this agent from partial agonism to functional selectivity. From the induction of early genes to modulation of scaffolding proteins and activation of transcription factors, aripiprazole has been shown to affect multiple cellular pathways and several cortical and subcortical neurotransmitter circuitries. Growing evidence shows that, beyond the consequences of D2R occupancy, aripiprazole has a unique neurobiology among available antipsychotics. The effect of chronic administration of aripiprazole on D2R affinity state and number has been especially highlighted, with relevant translational implications for long-term treatment of psychosis. The hypothesized effects of aripiprazole on cell-protective mechanisms and neurite growth, as well as the differential effects on intracellular pathways [i.e. extracellular signal-regulated kinase (ERK)] compared with full D2R antagonists, suggest further exploration of these targets by novel and future biased ligand compounds. This review aims to recapitulate the main neurobiological effects of aripiprazole and discuss the potential implications for upcoming improvements in schizophrenia therapy based on dopamine modulation beyond D2R antagonism.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Unit of Treatment Resistant Psychosis, Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine of Napoli "Federico II", Via Pansini, 5, Edificio n.18, 3rd floor, 80131, Naples, Italy.
| | - Carmine Tomasetti
- Unit of Treatment Resistant Psychosis, Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine of Napoli "Federico II", Via Pansini, 5, Edificio n.18, 3rd floor, 80131, Naples, Italy
| | - Felice Iasevoli
- Unit of Treatment Resistant Psychosis, Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine of Napoli "Federico II", Via Pansini, 5, Edificio n.18, 3rd floor, 80131, Naples, Italy
| |
Collapse
|