1
|
Singh SB, Tiwari A, Katta MR, Kafle R, Ayubcha C, Patel KH, Bhattarai Y, Werner TJ, Alavi A, Revheim ME. The utility of PET imaging in depression. Front Psychiatry 2024; 15:1322118. [PMID: 38711875 PMCID: PMC11070570 DOI: 10.3389/fpsyt.2024.1322118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
This educational review article aims to discuss growing evidence from PET studies in the diagnosis and treatment of depression. PET has been used in depression to explore the neurotransmitters involved, the alterations in neuroreceptors, non-neuroreceptor targets (e.g., microglia and astrocytes), the severity and duration of the disease, the pharmacodynamics of various antidepressants, and neurobiological mechanisms of non-pharmacological therapies like psychotherapy, electroconvulsive therapy, and deep brain stimulation therapy, by showing changes in brain metabolism and receptor and non-receptor targets. Studies have revealed alterations in neurotransmitter systems such as serotonin, dopamine, GABA, and glutamate, which are linked to the pathophysiology of depression. Overall, PET imaging has furthered the neurobiological understanding of depression. Despite these advancements, PET findings have not yet led to significant changes in evidence-based practices. Addressing the reasons behind inconsistencies in PET imaging results, conducting large sample size studies with a more standardized methodological approach, and investigating further the genetic and neurobiological aspects of depression may better leverage PET imaging in future studies.
Collapse
Affiliation(s)
- Shashi B. Singh
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Atit Tiwari
- BP Koirala Institute of Health Sciences, Dharan, Nepal
| | | | - Riju Kafle
- Rhythm Neuropsychiatry Hospital and Research Center Pvt. Ltd, Lalitpur, Nepal
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, United States
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Krishna H. Patel
- Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Yash Bhattarai
- Case Western Reserve University/The MetroHealth System, Cleveland, OH, United States
| | - Thomas J. Werner
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Mona-Elisabeth Revheim
- The Intervention Center, Division of Technology and Innovation, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Hart XM, Spangemacher M, Defert J, Uchida H, Gründer G. Update Lessons from PET Imaging Part II: A Systematic Critical Review on Therapeutic Plasma Concentrations of Antidepressants. Ther Drug Monit 2024; 46:155-169. [PMID: 38287888 DOI: 10.1097/ftd.0000000000001142] [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: 06/15/2023] [Accepted: 07/29/2023] [Indexed: 01/31/2024]
Abstract
BACKGROUND Compared with antipsychotics, the relationship between antidepressant blood (plasma or serum) concentrations and target engagement is less well-established. METHODS We have discussed the literature on the relationship between plasma concentrations of antidepressant drugs and their target occupancy. Antidepressants reviewed in this work are citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, venlafaxine, duloxetine, milnacipran, tricyclic antidepressants (amitriptyline, nortriptyline, and clomipramine), bupropion, tranylcypromine, moclobemide, and vortioxetine. Four electronic databases were systematically searched. RESULTS We included 32 articles published 1996-2022. A strong relationship between serotonin transporter (SERT) occupancy and drug concentration is well established for selective serotonin reuptake inhibitors. Lower limits of recommended therapeutic reference ranges largely corroborate with the findings from positron emission tomography studies (80% SERT occupancy). Only a few novel studies have investigated alternative targets, that is, norepinephrine transporters (NETs), dopamine transporters (DATs), or monoamine oxidase A (MAO-A). For certain classes of drugs, positron emission tomography study data are inconclusive. Low DAT occupancy after bupropion treatment speculates its discussed mechanism of action. For MAO inhibitors, a correlation between drug concentration and MAO-A occupancy could not be established. CONCLUSIONS Neuroimaging studies are critical in TDM-guided therapy for certain antidepressants, whereas for bupropion and MAO inhibitors, the available evidence offers no further insight. Evidence for selective serotonin reuptake inhibitors is strong and justifies a titration toward suggested ranges. For SNRIs, duloxetine, and venlafaxine, NETs are sufficiently occupied, well above the SERT efficacy threshold. For these drugs, a titration toward higher concentrations (within the recommended range) should be considered in case of no response at lower concentrations.
Collapse
Affiliation(s)
- Xenia M Hart
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; and
| | - Moritz Spangemacher
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Julie Defert
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; and
| | - Gerhard Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
3
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Pinto B, Conde T, Domingues I, Domingues MR. Adaptation of Lipid Profiling in Depression Disease and Treatment: A Critical Review. Int J Mol Sci 2022; 23:ijms23042032. [PMID: 35216147 PMCID: PMC8874755 DOI: 10.3390/ijms23042032] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
Major depressive disorder (MDD), also called depression, is a serious disease that impairs the quality of life of patients and has a high incidence, affecting approximately 3.8% of the world population. Its diagnosis is very subjective and is not supported by measurable biomarkers mainly due to the lack of biochemical markers. Recently, disturbance of lipid profiling has been recognized in MDD, in animal models of MDD or in depressed patients, which may contribute to unravel the etiology of the disease and find putative new biomarkers, for a diagnosis or for monitoring the disease and therapeutics outcomes. In this review, we provide an overview of current knowledge of lipidomics analysis, both in animal models of MDD (at the brain and plasma level) and in humans (in plasma and serum). Furthermore, studies of lipidomics analyses after antidepressant treatment in rodents (in brain, plasma, and serum), in primates (in the brain) and in humans (in plasma) were reviewed and give evidence that antidepressants seem to counteract the modification seen in lipids in MDD, giving some evidence that certain altered lipid profiles could be useful MDD biomarkers for future precision medicine.
Collapse
Affiliation(s)
- Bruno Pinto
- Centre for Environmental and Marine Studies, CESAM, Department of Chemistry, Santiago University Campus, University of Aveiro, 3810-193 Aveiro, Portugal; (B.P.); (T.C.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, Santiago University Campus, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Tiago Conde
- Centre for Environmental and Marine Studies, CESAM, Department of Chemistry, Santiago University Campus, University of Aveiro, 3810-193 Aveiro, Portugal; (B.P.); (T.C.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, Santiago University Campus, University of Aveiro, 3810-193 Aveiro, Portugal
- Institute of Biomedicine—iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Inês Domingues
- Centre for Environmental and Marine Studies, CESAM, Department of Biology, Santiago University Campus, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - M. Rosário Domingues
- Centre for Environmental and Marine Studies, CESAM, Department of Chemistry, Santiago University Campus, University of Aveiro, 3810-193 Aveiro, Portugal; (B.P.); (T.C.)
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, Santiago University Campus, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence:
| |
Collapse
|
5
|
Sørensen A, Ruhé HG, Munkholm K. The relationship between dose and serotonin transporter occupancy of antidepressants-a systematic review. Mol Psychiatry 2022; 27:192-201. [PMID: 34548628 PMCID: PMC8960396 DOI: 10.1038/s41380-021-01285-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/11/2021] [Accepted: 08/25/2021] [Indexed: 11/09/2022]
Abstract
Brain imaging techniques enable the visualization of serotonin transporter (SERT) occupancy as a measure of the proportion of SERT blocked by an antidepressant at a given dose. We aimed to systematically review the evidence on the relationship between antidepressant dose and SERT occupancy. We searched PubMed and Embase (last search 20 May 2021) for human in vivo, within-subject PET, or SPECT studies measuring SERT occupancy at any dose of any antidepressant with highly selective radioligands ([11C]-DASB, [123I]-ADAM, and [11C]-MADAM). We summarized and visualized the dose-occupancy relationship for antidepressants across studies, overlaying the plots with a curve based on predicted values of a standard 2-parameter Michaelis-Menten model fitted using the observed data. We included seventeen studies of 10 different SSRIs, SNRIs, and serotonin modulators comprising a total of 294 participants, involving 309 unique occupancy measures. Overall, following the Michaelis-Menten equation, SERT occupancy increased with a higher dose in a hyperbolic relationship, with occupancy increasing rapidly at lower doses and reaching a plateau at approximately 80% at the usual minimum recommended dose. All the studies were small, only a few investigated the same antidepressant, dose, and brain region, and few reported information on factors that may influence SERT occupancy. The hyperbolic dose-occupancy relationship may provide mechanistic insight of relevance to the limited clinical benefit of dose-escalation in antidepressant treatment and the potential emergence of withdrawal symptoms. The evidence is limited by non-transparent reporting, lack of standardized methods, small sample sizes, and short treatment duration. Future studies should standardize the imaging and reporting procedures, measure occupancy at lower antidepressant doses, and investigate the moderators of the dose-occupancy relationship.
Collapse
Affiliation(s)
- Anders Sørensen
- Nordic Cochrane Centre, Rigshospitalet, Copenhagen, Denmark.
| | - Henricus G. Ruhé
- grid.10417.330000 0004 0444 9382Department of Psychiatry, Radboudumc, Nijmegen, The Netherlands ,grid.5590.90000000122931605Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, The Netherlands
| | - Klaus Munkholm
- grid.10825.3e0000 0001 0728 0170Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark ,grid.7143.10000 0004 0512 5013Open Patient data Exploratory Network (OPEN), Odense University Hospital, Odense, Denmark
| |
Collapse
|
6
|
Eap CB, Gründer G, Baumann P, Ansermot N, Conca A, Corruble E, Crettol S, Dahl ML, de Leon J, Greiner C, Howes O, Kim E, Lanzenberger R, Meyer JH, Moessner R, Mulder H, Müller DJ, Reis M, Riederer P, Ruhe HG, Spigset O, Spina E, Stegman B, Steimer W, Stingl J, Suzen S, Uchida H, Unterecker S, Vandenberghe F, Hiemke C. Tools for optimising pharmacotherapy in psychiatry (therapeutic drug monitoring, molecular brain imaging and pharmacogenetic tests): focus on antidepressants. World J Biol Psychiatry 2021; 22:561-628. [PMID: 33977870 DOI: 10.1080/15622975.2021.1878427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objectives: More than 40 drugs are available to treat affective disorders. Individual selection of the optimal drug and dose is required to attain the highest possible efficacy and acceptable tolerability for every patient.Methods: This review, which includes more than 500 articles selected by 30 experts, combines relevant knowledge on studies investigating the pharmacokinetics, pharmacodynamics and pharmacogenetics of 33 antidepressant drugs and of 4 drugs approved for augmentation in cases of insufficient response to antidepressant monotherapy. Such studies typically measure drug concentrations in blood (i.e. therapeutic drug monitoring) and genotype relevant genetic polymorphisms of enzymes, transporters or receptors involved in drug metabolism or mechanism of action. Imaging studies, primarily positron emission tomography that relates drug concentrations in blood and radioligand binding, are considered to quantify target structure occupancy by the antidepressant drugs in vivo. Results: Evidence is given that in vivo imaging, therapeutic drug monitoring and genotyping and/or phenotyping of drug metabolising enzymes should be an integral part in the development of any new antidepressant drug.Conclusions: To guide antidepressant drug therapy in everyday practice, there are multiple indications such as uncertain adherence, polypharmacy, nonresponse and/or adverse reactions under therapeutically recommended doses, where therapeutic drug monitoring and cytochrome P450 genotyping and/or phenotyping should be applied as valid tools of precision medicine.
Collapse
Affiliation(s)
- C B Eap
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Lausanne, Switzerland, Geneva, Switzerland
| | - G Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - P Baumann
- Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - N Ansermot
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - A Conca
- Department of Psychiatry, Health Service District Bolzano, Bolzano, Italy.,Department of Child and Adolescent Psychiatry, South Tyrolean Regional Health Service, Bolzano, Italy
| | - E Corruble
- INSERM CESP, Team ≪MOODS≫, Service Hospitalo-Universitaire de Psychiatrie, Universite Paris Saclay, Le Kremlin Bicetre, France.,Service Hospitalo-Universitaire de Psychiatrie, Hôpital Bicêtre, Assistance Publique Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - S Crettol
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - M L Dahl
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - J de Leon
- Eastern State Hospital, University of Kentucky Mental Health Research Center, Lexington, KY, USA
| | - C Greiner
- Bundesinstitut für Arzneimittel und Medizinprodukte, Bonn, Germany
| | - O Howes
- King's College London and MRC London Institute of Medical Sciences (LMS)-Imperial College, London, UK
| | - E Kim
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - R Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - J H Meyer
- Campbell Family Mental Health Research Institute, CAMH and Department of Psychiatry, University of Toronto, Toronto, Canada
| | - R Moessner
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - H Mulder
- Department of Clinical Pharmacy, Wilhelmina Hospital Assen, Assen, The Netherlands.,GGZ Drenthe Mental Health Services Drenthe, Assen, The Netherlands.,Department of Pharmacotherapy, Epidemiology and Economics, Department of Pharmacy and Pharmaceutical Sciences, University of Groningen, Groningen, The Netherlands.,Department of Psychiatry, Interdisciplinary Centre for Psychopathology and Emotion Regulation, University of Groningen, Groningen, The Netherlands
| | - D J Müller
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - M Reis
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Clinical Chemistry and Pharmacology, Skåne University Hospital, Lund, Sweden
| | - P Riederer
- Center of Mental Health, Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany.,Department of Psychiatry, University of Southern Denmark Odense, Odense, Denmark
| | - H G Ruhe
- Department of Psychiatry, Radboudumc, Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| | - O Spigset
- Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - E Spina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - B Stegman
- Institut für Pharmazie der Universität Regensburg, Regensburg, Germany
| | - W Steimer
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Munich, Germany
| | - J Stingl
- Institute for Clinical Pharmacology, University Hospital of RWTH Aachen, Germany
| | - S Suzen
- Department of Toxicology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - H Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - S Unterecker
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - F Vandenberghe
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Center for Psychiatric Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - C Hiemke
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
7
|
Arakawa R, Takano A, Halldin C. PET technology for drug development in psychiatry. Neuropsychopharmacol Rep 2020; 40:114-121. [PMID: 32463584 PMCID: PMC7722687 DOI: 10.1002/npr2.12084] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/05/2019] [Accepted: 10/18/2019] [Indexed: 12/14/2022] Open
Abstract
Positron emission tomography (PET) is a non‐invasive imaging method to measure the molecule in vivo. PET imaging can evaluate the central nervous system drugs as target engagement in the human brain. For antipsychotic drugs, adequate dopamine D2 receptor occupancy (“therapeutic window”) is reported to be from 65%‐70% to 80% to achieve the antipsychotic effect without extrapyramidal symptoms. For antidepressants, the clinical threshold of serotonin transporter (5‐HTT) occupancy is reported to be 70%‐80% although the relation between the side effect and 5‐HTT occupancy has not yet been established. Evaluation of norepinephrine transporter (NET) occupancy for antidepressant is ongoing as adequate PET radioligands for NET were developed recently. Measurement of the target occupancy has been a key element to evaluate the in vivo target engagement of the drugs. In order to evaluate new drug targets for disease conditions such as negative symptoms/cognitive impairment of schizophrenia and treatment‐resistant depression, new PET radioligands need to be developed concurrently with the drug development. PET imaging can evaluate the central nervous system drugs as target engagement in the human brain. The uptake of [11C]raclopride for dopamine D2 receptors decreased from (A) baseline to (B) antipsychotic administration conditions.![]()
Collapse
Affiliation(s)
- Ryosuke Arakawa
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Akihiro Takano
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.,Takeda Development Center Japan, Takeda Pharmaceutical Company Limited, Osaka, Japan
| | - Christer Halldin
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
8
|
Recent advances in radiotracers targeting norepinephrine transporter: structural development and radiolabeling improvements. J Neural Transm (Vienna) 2020; 127:851-873. [PMID: 32274584 PMCID: PMC7223405 DOI: 10.1007/s00702-020-02180-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/21/2020] [Indexed: 12/13/2022]
Abstract
The norepinephrine transporter (NET) is a major target for the evaluation of the cardiac sympathetic nerve system in patients with heart failure and Parkinson's disease. It is also used in the therapeutic applications against certain types of neuroendocrine tumors, as exemplified by the clinically used 123/131I-MIBG as theranostic single-photon emission computed tomography (SPECT) agent. With the development of more advanced positron emission tomography (PET) technology, more radiotracers targeting NET have been reported, with superior temporal and spatial resolutions, along with the possibility of functional and kinetic analysis. More recently, fluorine-18-labelled NET tracers have drawn increasing attentions from researchers, due to their longer radiological half-life relative to carbon-11 (110 min vs. 20 min), reduced dependence on on-site cyclotrons, and flexibility in the design of novel tracer structures. In the heart, certain NET tracers provide integral diagnostic information on sympathetic innervation and the nerve status. In the central nervous system, such radiotracers can reveal NET distribution and density in pathological conditions. Most radiotracers targeting cardiac NET-function for the cardiac application consistent of derivatives of either norepinephrine or MIBG with its benzylguanidine core structure, e.g. 11C-HED and 18F-LMI1195. In contrast, all NET tracers used in central nervous system applications are derived from clinically used antidepressants. Lastly, possible applications of NET as selective tracers over organic cation transporters (OCTs) in the kidneys and other organs controlled by sympathetic nervous system will also be discussed.
Collapse
|
9
|
Item-based analysis of the effects of duloxetine in depression: a patient-level post hoc study. Neuropsychopharmacology 2020; 45:553-560. [PMID: 31521062 PMCID: PMC6969189 DOI: 10.1038/s41386-019-0523-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/12/2019] [Accepted: 09/03/2019] [Indexed: 11/13/2022]
Abstract
Oft-cited trial-level meta-analyses casting doubt on the usefulness of antidepressants have been based on re-analyses of to what extent the active drug has outperformed placebo in reducing the sum score of the Hamilton Depression Rating Scale (HDRS-17-sum) in clinical trials. Recent studies, however, suggest patient-level analyses of individual HDRS items to be more informative when assessing the efficacy of an antidepressant. To shed further light on both symptom-reducing and symptom-aggravating effects of a serotonin and noradrenaline reuptake inhibitor, duloxetine, when used for major depression in adults, we hence applied this approach to re-analyse data from 13 placebo-controlled trials. In addition, using patient-level data from 28 placebo-controlled trials of selective serotonin reuptake inhibitors (SSRIs), the response profile of duloxetine was compared to that of these drugs. Duloxetine induced a robust reduction in depressed mood that was not dependent on baseline severity and not caused by side-effects breaking the blind. A beneficial effect on depressed mood was at hand already after one week; when outcome was assessed using HDRS-17-sum as effect parameter, this early response was however masked by a concomitant deterioration with respect to adverse event-related items. No support for a suicide-provoking effect of duloxetine was obtained. The response profile of duloxetine was strikingly similar to that of the SSRIs. We conclude that the use of HDRS-17-sum as effect parameter underestimates the true efficacy and masks an early effect of duloxetine on core symptoms of depression. No support for major differences between duloxetine and SSRIs in clinical profile were obtained.
Collapse
|
10
|
De Donatis D, Florio V, Porcelli S, Saria A, Mercolini L, Serretti A, Conca A. Duloxetine plasma level and antidepressant response. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:127-132. [PMID: 30611837 DOI: 10.1016/j.pnpbp.2019.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/11/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Major Depressive Disorder (MDD) is associated with a high rate of inadequate treatment response, which is mainly due to the large inter-individual genetic variability in pharmacokinetic and pharmacodynamic targets of antidepressant drugs. Little is still known about the exact association between plasma level of first-line antidepressants and clinical response. This is particularly true for duloxetine, a dual serotonin and norepinephrine reuptake inhibitor recommended as first-line treatment for MDD. The aim of this study was to investigate the association between serum concentration of duloxetine (SCD) and antidepressant response (AR). METHODS 66 MDD patients treated with duloxetine 60 mg/day monotherapy were recruited in an outpatient setting and followed for three months. Hamilton Depression Rating Scale - 21 (HAMD-21) was administrated at baseline, at month 1, and at month 3 to assess AR. SCD was measured at steady state. Linear regression analysis and nonlinear least-squares regression were used to estimate association between SCD and AR. RESULTS SCD showed a high inter-individual variability in our sample, despite the duloxetine fixed oral dosage. We found a strong association between SCD and AR following a bell-shaped function at month 1 and at month 3. Nonetheless, within the recommended SCD range of 30-120 ng/mL a more linear correlation between SCD and AR was observed. DISCUSSION Our results suggest that for duloxetine the association between SCD and AR likely follows a bell-shaped quadratic function with poor AR at subtherapeutic SCD and progressive decrease of AR at higher SCD. The maximum antidepressant efficacy seems to require SCD values next to the highest recommended SCD (30-120 ng/mL), probably because of the optimal saturation of both serotonin and norepinephrine transporters. Thus, taking into account the observed high interindividual variability of SCD, our findings suggest that for MDD patients treated with duloxetine, SCD could be a useful tool to guide the treatment by optimizing the oral dosage in order to increase the AR rate.
Collapse
Affiliation(s)
- Domenico De Donatis
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | | | - Stefano Porcelli
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | - Alois Saria
- Experimental Psychiatry Unit, Medical University of Innsbruck, Innsbruck, Austria
| | - Laura Mercolini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy.
| | | |
Collapse
|
11
|
Shidahara Y, Natsume T, Awaga Y, Ogawa S, Yamoto K, Okamoto S, Hama A, Hayashi I, Takamatsu H, Magata Y. Distinguishing analgesic drugs from non-analgesic drugs based on brain activation in macaques with oxaliplatin-induced neuropathic pain. Neuropharmacology 2019; 149:204-211. [PMID: 30817933 DOI: 10.1016/j.neuropharm.2019.02.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 01/28/2019] [Accepted: 02/24/2019] [Indexed: 11/16/2022]
Abstract
The antineoplastic agent oxaliplatin is a first-line treatment for colorectal cancer. However, neuropathic pain, characterized by hypersensitivity to cold, emerges soon after treatment. In severe instances, dose reduction or curtailing treatment may be necessary. While a number of potential treatments for oxaliplatin-induced neuropathic pain have been proposed based on preclinical findings, few have demonstrated efficacy in randomized, placebo-controlled clinical studies. This failure could be related, in part, to the use of rodents as the primary preclinical species, as there are a number of distinctions in pain-related mechanisms between rodents and humans. Also, an indicator of preclinical pharmacological efficacy less subjective than behavioral endpoints that is translatable to clinical usage is lacking. Three days after oxaliplatin treatment in Macaca fascicularis, a significantly reduced response latency to cold (10 °C) water was observed, indicating cold hypersensitivity. Cold-evoked bilateral activation of the secondary somatosensory (SII) and insular (Ins) cortex was observed with functional magnetic resonance imaging. Duloxetine alleviated cold hypersensitivity and significantly attenuated activation in both SII and Ins. By contrast, neither clinically used analgesics pregabalin nor tramadol affected cold hypersensitivity and cold-evoked activation of SII and Ins. The current findings suggest that suppressing SII and Ins activation leads to antinociception, and, therefore, could be used as a non-behavioral indicator of analgesic efficacy in patients with oxaliplatin-induced neuropathic pain.
Collapse
Affiliation(s)
- Yuka Shidahara
- Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka Prefecture, Japan
| | - Takahiro Natsume
- Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka Prefecture, Japan
| | - Yūji Awaga
- Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka Prefecture, Japan
| | - Shin'ya Ogawa
- Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka Prefecture, Japan
| | - Kurumi Yamoto
- Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka Prefecture, Japan
| | - Shinichi Okamoto
- Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka Prefecture, Japan
| | - Aldric Hama
- Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka Prefecture, Japan.
| | - Ikuo Hayashi
- Hamamatsu Pharma Research, USA, Inc., San Diego, CA, USA
| | | | - Yasuhiro Magata
- Department of Molecular Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka Prefecture, Japan
| |
Collapse
|
12
|
Arakawa R, Stenkrona P, Takano A, Svensson J, Andersson M, Nag S, Asami Y, Hirano Y, Halldin C, Lundberg J. Venlafaxine ER Blocks the Norepinephrine Transporter in the Brain of Patients with Major Depressive Disorder: a PET Study Using [18F]FMeNER-D2. Int J Neuropsychopharmacol 2019; 22:278-285. [PMID: 30649319 PMCID: PMC6441126 DOI: 10.1093/ijnp/pyz003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/21/2018] [Accepted: 01/09/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The in vivo binding of clinical dose of venlafaxine on norepinephrine transporter has been questioned because venlafaxine has higher in vitro affinity to serotonin transporter than that to norepinephrine transporter. Although serotonin transporter occupancy of clinically relevant doses of venlafaxine has been reported, there has been no report of norepinephrine transporter occupancy in the human brain. METHODS This was an open-label, single center, exploratory positron emission tomography study. Twelve major depressive disorder patients who had responded to venlafaxine extended-release and 9 control subjects were recruited. Each subject participated in one positron emission tomography measurement with [18F]FMeNER-D2. Binding potential in brain was quantified by the area under the curve ratio method with thalamus as target and white matter as reference regions. The difference of binding potential values between control and patient groups divided to 2 dose ranges were evaluated. Norepinephrine transporter occupancy (%) for all the major depressive disorder patients was calculated using mean binding potential of control subjects as baseline. The relationships between dose or plasma concentration of total active moiety and occupancies of norepinephrine transporter were also estimated. RESULTS The binding potential of the patient group with 150 to 300 mg/d was significantly lower than that in the control subjects group (P = .0004 < .05/2). The norepinephrine transporter occupancy (8-61%) increased in a dose-dependent manner although a clear difference beyond 150 mg/d was not observed. CONCLUSIONS This study demonstrates that clinically relevant doses of venlafaxine extended-release block the norepinephrine transporter of the major depressive disorder patient's brain. The data support the notion that the antidepressant effect of venlafaxine involves a combination of serotonin transporter and norepinephrine transporter blockades.
Collapse
Affiliation(s)
- Ryosuke Arakawa
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden,Correspondence: Ryosuke Arakawa, MD, PhD, Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden. Post address: Karolinska University Hospital Solna, R5:02, SE-17176 Stockholm, Sweden ()
| | - Per Stenkrona
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Akihiro Takano
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Jonas Svensson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Max Andersson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Sangram Nag
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Yuko Asami
- Central Nervous System, Medical Affairs, Pfizer Essential Health, Pfizer Japan Inc., Tokyo, Japan
| | - Yoko Hirano
- Central Nervous System, Medical Affairs, Pfizer Essential Health, Pfizer Japan Inc., Tokyo, Japan
| | - Christer Halldin
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Johan Lundberg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
13
|
Jesulola E, Micalos P, Baguley IJ. Understanding the pathophysiology of depression: From monoamines to the neurogenesis hypothesis model - are we there yet? Behav Brain Res 2017; 341:79-90. [PMID: 29284108 DOI: 10.1016/j.bbr.2017.12.025] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 02/07/2023]
Abstract
A number of factors (biogenic amine deficiency, genetic, environmental, immunologic, endocrine factors and neurogenesis) have been identified as mechanisms which provide unitary explanations for the pathophysiology of depression. Rather than a unitary construct, the combination and linkage of these factors have been implicated in the pathogenesis of depression. That is, environmental stressors and heritable genetic factors acting through immunologic and endocrine responses initiate structural and functional changes in many brain regions, resulting in dysfunctional neurogenesis and neurotransmission which then manifest as a constellation of symptoms which present as depression.
Collapse
Affiliation(s)
- Emmanuel Jesulola
- Paramedicine Discipline, Charles Sturt University, Bathurst Campus, NSW Australia.
| | - Peter Micalos
- Paramedicine Discipline, Charles Sturt University, Bathurst Campus, NSW Australia
| | - Ian J Baguley
- Brain Injury Rehabilitation Service, Westmead Hospital, Hawkesbury Rd, Wentworthville, NSW Australia
| |
Collapse
|