1
|
Bartlett EA, Yttredahl AA, Boldrini M, Tyrer AE, Hill KR, Ananth MR, Milak MS, Oquendo MA, Mann JJ, DeLorenzo C, Parsey RV. In vivo serotonin 1A receptor hippocampal binding potential in depression and reported childhood adversity. Eur Psychiatry 2023; 66:e17. [PMID: 36691786 PMCID: PMC9970152 DOI: 10.1192/j.eurpsy.2023.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Reported childhood adversity (CA) is associated with development of depression in adulthood and predicts a more severe course of illness. Although elevated serotonin 1A receptor (5-HT1AR) binding potential, especially in the raphe nuclei, has been shown to be a trait associated with major depression, we did not replicate this finding in an independent sample using the partial agonist positron emission tomography tracer [11C]CUMI-101. Evidence suggests that CA can induce long-lasting changes in expression of 5-HT1AR, and thus, a history of CA may explain the disparate findings. METHODS Following up on our initial report, 28 unmedicated participants in a current depressive episode (bipolar n = 16, unipolar n = 12) and 19 non-depressed healthy volunteers (HVs) underwent [11C]CUMI-101 imaging to quantify 5-HT1AR binding potential. Participants in a depressive episode were stratified into mild/moderate and severe CA groups via the Childhood Trauma Questionnaire. We hypothesized higher hippocampal and raphe nuclei 5-HT1AR with severe CA compared with mild/moderate CA and HVs. RESULTS There was a group-by-region effect (p = 0.011) when considering HV, depressive episode mild/moderate CA, and depressive episode severe CA groups, driven by significantly higher hippocampal 5-HT1AR binding potential in participants in a depressive episode with severe CA relative to HVs (p = 0.019). Contrary to our hypothesis, no significant binding potential differences were detected in the raphe nuclei (p-values > 0.05). CONCLUSIONS With replication in larger samples, elevated hippocampal 5-HT1AR binding potential may serve as a promising biomarker through which to investigate the neurobiological link between CA and depression.
Collapse
Affiliation(s)
- Elizabeth A Bartlett
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York10032, USA.,Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York10032, USA
| | - Ashley A Yttredahl
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York10032, USA.,Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York10032, USA
| | - Maura Boldrini
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York10032, USA
| | - Andrea E Tyrer
- Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY11794, USA.,Clinical Genetics Research Program, Centre for Addiction and Mental Health, University of Toronto, Toronto, OntarioM5S, Canada
| | - Kathryn R Hill
- Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY11794, USA
| | - Mala R Ananth
- National Institute of Neurological Disorders and Stroke, National Institute of Health, Bethesda, Maryland20892, USA
| | - Matthew S Milak
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York10032, USA.,Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York10032, USA
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania19104, USA
| | - J John Mann
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York10032, USA.,Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York10032, USA.,Department of Radiology, Columbia University, New York, New York10027, USA
| | - Christine DeLorenzo
- Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY11794, USA.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York11794, USA
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook Medicine, Stony Brook, NY11794, USA.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York11794, USA.,Department of Radiology, Stony Brook University, Stony Brook, New York11794, USA
| |
Collapse
|
2
|
Ultrasound assisted one-pot synthesis and preliminary in vitro studies of salicylamide arylpiperazines as dual 5-HT1A/5-HT7 ligands. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
3
|
Fu H, Rong J, Chen Z, Zhou J, Collier T, Liang SH. Positron Emission Tomography (PET) Imaging Tracers for Serotonin Receptors. J Med Chem 2022; 65:10755-10808. [PMID: 35939391 DOI: 10.1021/acs.jmedchem.2c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) and 5-HT receptors (5-HTRs) have crucial roles in various neuropsychiatric disorders and neurodegenerative diseases, making them attractive diagnostic and therapeutic targets. Positron emission tomography (PET) is a noninvasive nuclear molecular imaging technique and is an essential tool in clinical diagnosis and drug discovery. In this context, numerous PET ligands have been developed for "visualizing" 5-HTRs in the brain and translated into human use to study disease mechanisms and/or support drug development. Herein, we present a comprehensive repertoire of 5-HTR PET ligands by focusing on their chemotypes and performance in PET imaging studies. Furthermore, this Perspective summarizes recent 5-HTR-focused drug discovery, including biased agonists and allosteric modulators, which would stimulate the development of more potent and subtype-selective 5-HTR PET ligands and thus further our understanding of 5-HTR biology.
Collapse
Affiliation(s)
- Hualong Fu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Zhen Chen
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Jingyin Zhou
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Thomas Collier
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, Massachusetts 02114, United States.,Department of Radiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
4
|
Mangeant R, Dubost E, Cailly T, Collot V. Radiotracers for the Central Serotoninergic System. Pharmaceuticals (Basel) 2022; 15:571. [PMID: 35631397 PMCID: PMC9143978 DOI: 10.3390/ph15050571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
This review lists the most important radiotracers described so far for imaging the central serotoninergic system. Single-photon emission computed tomography and positron emission tomography radiotracers are reviewed and critically discussed for each receptor.
Collapse
Affiliation(s)
- Reynald Mangeant
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| | - Emmanuelle Dubost
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| | - Thomas Cailly
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
- UNICAEN, IMOGERE, Normandie Univ., 14000 Caen, France
- CHU Côte de Nacre, Department of Nuclear Medicine, 14000 Caen, France
| | - Valérie Collot
- Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), UNICAEN, Normandie Univ., 14000 Caen, France; (R.M.); (E.D.)
- Institut Blood and Brain @ Caen Normandie (BB@C), Boulevard Henri Becquerel, 14000 Caen, France
| |
Collapse
|
5
|
Melhem NM, Zhong Y, Miller JM, Zanderigo F, Ogden RT, Sublette ME, Newell M, Burke A, Keilp JG, Lesanpezeshki M, Bartlett E, Brent DA, Mann JJ. Brain 5-HT1A Receptor PET Binding, Cortisol Responses to Stress, and the Familial Transmission of Suicidal Behavior. Int J Neuropsychopharmacol 2021; 25:36-45. [PMID: 34555145 PMCID: PMC8756092 DOI: 10.1093/ijnp/pyab060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/22/2021] [Accepted: 10/04/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The serotonin 1A (5-HT1A) receptor has been implicated in depression and suicidal behavior. Lower resting cortisol levels are associated with higher 5-HT1A receptor binding, and both differentiate suicide attempters with depression. However, it is not clear whether 5-HT1A receptor binding and cortisol responses to stress are related to familial risk and resilience for suicidal behavior. METHODS [11C]CUMI-101 positron emission tomography imaging to quantify regional brain 5-HT1A receptor binding was conducted in individuals considered to be at high risk for mood disorder or suicidal behavior on the basis of having a first- or second-degree relative(s) with an early onset mood disorder and history of suicidal behavior. These high-risk individuals were subdivided into the following groups: high risk resilient having no mood disorder or suicidal behavior (n = 29); high risk with mood disorder and no suicidal behavior history (n = 31); and high risk with mood disorder and suicidal behavior (n = 25). Groups were compared with healthy volunteers without a family history of mood disorder or suicidal behavior (n = 34). Participants underwent the Trier Social Stress Task (TSST). All participants were free from psychotropic medications at the time of the TSST and PET scanning. RESULTS We observed no group differences in 5-HT1A receptor binding considering all regions simultaneously, nor did we observe heterogeneity of the effect of group across regions. These results were similar across outcome measures (BPND for all participants and BPp in a subset of the sample) and definitions of regions of interest (ROIs; standard or serotonin system-specific ROIs). We also found no group differences on TSST outcomes. Within the high risk with mood disorder and suicidal behavior group, lower BPp binding (β = -0.084, SE = 0.038, P = .048) and higher cortisol reactivity to stress (β = 9.25, 95% CI [3.27,15.23], P = .004) were associated with higher lethality attempts. There were no significant relationships between 5-HT1A binding and cortisol outcomes. CONCLUSIONS 5-HT1A receptor binding in ROIs was not linked to familial risk or resilience protecting against suicidal behavior or mood disorder although it may be related to lethality of suicide attempt. Future studies are needed to better understand the biological mechanisms implicated in familial risk for suicidal behavior and how hypothalamic-pituitary-adrenal axis function influences such risk.
Collapse
Affiliation(s)
- Nadine M Melhem
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Correspondence: Nadine Melhem, PhD, 3811 O’Hara Street, Pittsburgh, PA 15213, USA ()
| | - Yongqi Zhong
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Graduate School of Public Health, University of Pittsburgh
| | - Jeffrey M Miller
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Francesca Zanderigo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - R Todd Ogden
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York, United States
| | - M Elizabeth Sublette
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Madison Newell
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
| | - Ainsley Burke
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - John G Keilp
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Mohammad Lesanpezeshki
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - Elizabeth Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| | - David A Brent
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - J John Mann
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, United States
- Department of Psychiatry, Columbia University, New York, New York, United States
| |
Collapse
|
6
|
Lacivita E, Niso M, Mastromarino M, Garcia Silva A, Resch C, Zeug A, Loza MI, Castro M, Ponimaskin E, Leopoldo M. Knowledge-Based Design of Long-Chain Arylpiperazine Derivatives Targeting Multiple Serotonin Receptors as Potential Candidates for Treatment of Autism Spectrum Disorder. ACS Chem Neurosci 2021; 12:1313-1327. [PMID: 33792287 DOI: 10.1021/acschemneuro.0c00647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a group of neurodevelopmental disorders characterized by core symptoms such as impaired social interaction and communication, repetitive and stereotyped behaviors, and restricted interests. To date, there are no effective treatments for these core symptoms. Several studies have shown that the brain serotonin (5-HT) neurotransmission system is altered in both ASD patients and animal models of the disease. Multiple pieces of evidence suggest that targeting 5-HT receptors may treat the core symptoms of ASD and associated intellectual disabilities. In fact, stimulation of the 5-HT1A receptor reduces repetitive and restricted behaviors; blockade of the 5-HT2A receptor reduces both learning deficits and repetitive behavior, and activation of the 5-HT7 receptor improves cognitive performances and reduces repetitive behavior. On such a basis, we have designed novel arylpiperazine derivatives pursuing unprecedently reported activity profiles: dual 5-HT7/5-HT1A receptor agonist properties and mixed 5-HT7 agonist/5-HT1A agonist/5-HT2A antagonist properties. Seventeen new compounds were synthesized and tested in radioligand binding assay at the target receptors. We have identified the dual 5-HT1AR/5-HT7R agonists 8c and 29 and the mixed 5-HT1AR agonist/5-HT7R agonist/5-HT2AR antagonist 20b. These compounds are metabolically stable in vitro and have suitable central nervous system druglike properties.
Collapse
Affiliation(s)
- Enza Lacivita
- Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| | - Mauro Niso
- Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| | - Margherita Mastromarino
- Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| | - Andrea Garcia Silva
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
| | - Cibell Resch
- Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
| | - María I. Loza
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
| | - Marián Castro
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS). Universidade de Santiago de Compostela. Avda. de Barcelona, s/n, 15782 Santiago de Compostela, Spain
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, 30625 Hannover, Germany
| | - Marcello Leopoldo
- Dipartimento di Farmacia−Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| |
Collapse
|
7
|
Narayanaswami V, Tong J, Fiorino F, Severino B, Sparaco R, Magli E, Giordano F, Bloomfield PM, Prabhakaran J, Mann JJ, Vasdev N, Dahl K, Kumar JSD. Synthesis, in vitro and in vivo evaluation of 11C-O-methylated arylpiperazines as potential serotonin 1A (5-HT 1A) receptor antagonist radiotracers. EJNMMI Radiopharm Chem 2020; 5:13. [PMID: 32430632 PMCID: PMC7237647 DOI: 10.1186/s41181-020-00096-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/23/2020] [Indexed: 01/23/2023] Open
Abstract
Background Serotonin 1A (5-HT1A) receptors are implicated in the pathogenesis of several psychiatric and neurodegenerative disorders motivating the development of suitable radiotracers for in vivo positron emission tomography (PET) neuroimaging. The gold standard PET imaging agent for this target is [carbonyl-11C]WAY-100635, labeled via a technically challenging multi-step reaction that has limited its widespread use. While several antagonist and agonist-based PET radiotracers for 5-HT 1A receptors have been developed, their clinical translation has been hindered by methodological challenges and/or and non-specific binding. As a result, there is continued interest in the development of new and more selective 5-HT1A PET tracers having a relatively easier and reliable radiosynthesis process for routine production and with favorable metabolism to facilitate tracer-kinetic modeling. The purpose of the current study was to develop and characterize a radioligand with suitable characteristics for imaging 5-HT1A receptors in the brain. The current study reports the in vitro characterization and radiosyntheses of three candidate 5-HT1A receptor antagonists, DF-100 (1), DF-300 (2) and DF-400 (3), to explore their suitability as potential PET radiotracers. Results Syntheses of 1–3 and corresponding precursors for radiolabeling were achieved from isonicotinic, picolinic acid or picolino nitrile. In vitro binding studies demonstrated nanomolar affinity of the compounds for 5-HT1A receptors. Binding of 1–3 for other biogenic amines, neurotransmitter receptors, and transporters was negligible with the exception of moderate affinities for α1-adrenergic receptors (4–6-fold less potent than that for 5-HT1A receptor). Radioligands [11C]1–3 were efficiently prepared by 11C-O-methylation of the corresponding phenolic precursor in non-decay corrected radiochemical yields of 7–11% with > 99% chemical and radiochemical purities. Dynamic PET studies in rats demonstrated negligible brain uptake of [11C]1 and [11C]2. In contrast, significant brain uptake of [11C]3 was observed with an early peak SUV of 4–5. However, [11C]3 displayed significant off-target binding attributed to α1-adrenergic receptors based on regional distribution (thalamus>hippocampus) and blocking studies. Conclusion Despite efficient radiolabeling, results from PET imaging experiments limit the application of [11C]3 for in vivo quantification of 5-HT1A receptors. Nevertheless, derivatives of compound 3 may provide a scaffold for alternative PET radiotracers with improved selectivity for 5-HT 1A receptors or α1-adrenergic receptors.
Collapse
Affiliation(s)
- Vidya Narayanaswami
- Azrieli Centre for Neuro-Radiochemistry, Research Imaging Centre & Preclinical Imaging, Centre for Addiction and Mental Health, Toronto, Ontario, M5T-1R8, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry, Research Imaging Centre & Preclinical Imaging, Centre for Addiction and Mental Health, Toronto, Ontario, M5T-1R8, Canada
| | - Ferdinando Fiorino
- Department of Pharmacy, University of Naples, Via D. Montesano, 49, 8013, Naples, Italy
| | - Beatrice Severino
- Department of Pharmacy, University of Naples, Via D. Montesano, 49, 8013, Naples, Italy
| | - Rosa Sparaco
- Department of Pharmacy, University of Naples, Via D. Montesano, 49, 8013, Naples, Italy
| | - Elisa Magli
- Department of Pharmacy, University of Naples, Via D. Montesano, 49, 8013, Naples, Italy
| | - Flavia Giordano
- Department of Pharmacy, University of Naples, Via D. Montesano, 49, 8013, Naples, Italy
| | - Peter M Bloomfield
- Azrieli Centre for Neuro-Radiochemistry, Research Imaging Centre & Preclinical Imaging, Centre for Addiction and Mental Health, Toronto, Ontario, M5T-1R8, Canada
| | - Jaya Prabhakaran
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA
| | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA.,Department of Psychiatry, Columbia University Medical Center, New York, USA
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Research Imaging Centre & Preclinical Imaging, Centre for Addiction and Mental Health, Toronto, Ontario, M5T-1R8, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, M5T-1R8, Canada
| | - Kenneth Dahl
- Azrieli Centre for Neuro-Radiochemistry, Research Imaging Centre & Preclinical Imaging, Centre for Addiction and Mental Health, Toronto, Ontario, M5T-1R8, Canada.
| | - J S Dileep Kumar
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, USA.
| |
Collapse
|
8
|
Pillai RLI, Bartlett EA, Ananth MR, Zhu C, Yang J, Hajcak G, Parsey RV, DeLorenzo C. Examining the underpinnings of loudness dependence of auditory evoked potentials with positron emission tomography. Neuroimage 2020; 213:116733. [PMID: 32169543 DOI: 10.1016/j.neuroimage.2020.116733] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 11/30/2022] Open
Abstract
Loudness dependence of auditory evoked potentials (LDAEP) has long been considered to reflect central basal serotonin transmission. However, the relationship between LDAEP and individual serotonin receptors and transporters has not been fully explored in humans and may involve other neurotransmitter systems. To examine LDAEP's relationship with the serotonin system, we performed PET using serotonin-1A (5-HT1A) imaging via [11C]CUMI-101 and serotonin transporter (5-HTT) imaging via [11C]DASB on a mixed sample of healthy controls (n = 4: 4 females, 0 males), patients with unipolar (MDD, n = 11: 4 females, 7 males) and bipolar depression (BD, n = 8: 4 females, 4 males). On these same participants, we also performed electroencephalography (EEG) within a week of PET scanning, using 1000 Hz tones of varying intensity to evoke LDAEP. We then evaluated the relationship between LDAEP and 5-HT1A or 5-HTT binding in both the raphe (5-HT1A)/midbrain (5-HTT) areas and in the temporal cortex. We found that LDAEP was significantly correlated with 5-HT1A positively and with 5-HTT negatively in the temporal cortex (p < 0.05), but not correlated with either in midbrain or raphe. In males only, exploratory analysis showed multiple regions in which LDAEP significantly correlated with 5-HT1A throughout the brain; we did not find this with 5-HTT. This multimodal study partially validates preclinical models of a serotonergic influence on LDAEP. Replication in larger samples is necessary to further clarify our understanding of the role of serotonin in perception of auditory tones.
Collapse
Affiliation(s)
| | - Elizabeth A Bartlett
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, United States
| | - Mala R Ananth
- Department of Psychiatry, Stony Brook University, United States
| | - Chencan Zhu
- Department of Applied Mathematics and Statistics, Stony Brook University, United States
| | - Jie Yang
- Department of Family, Population, and Preventive Medicine, Stony Brook University, United States
| | - Greg Hajcak
- Department of Biomedical Sciences and Psychology, Florida State University, United States
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook University, United States
| | - Christine DeLorenzo
- Department of Psychiatry, Stony Brook University, United States; Department of Biomedical Engineering, Stony Brook University, United States
| |
Collapse
|
9
|
An improved synthesis of the 5-HT1A receptor agonist Eptapirone free base. CHEMICAL PAPERS 2019. [DOI: 10.1007/s11696-019-00685-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
10
|
Shalgunov V, van Waarde A, Booij J, Michel MC, Dierckx RAJO, Elsinga PH. Hunting for the high-affinity state of G-protein-coupled receptors with agonist tracers: Theoretical and practical considerations for positron emission tomography imaging. Med Res Rev 2018; 39:1014-1052. [PMID: 30450619 PMCID: PMC6587759 DOI: 10.1002/med.21552] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/02/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
Abstract
The concept of the high‐affinity state postulates that a certain subset of G‐protein‐coupled receptors is primarily responsible for receptor signaling in the living brain. Assessing the abundance of this subset is thus potentially highly relevant for studies concerning the responses of neurotransmission to pharmacological or physiological stimuli and the dysregulation of neurotransmission in neurological or psychiatric disorders. The high‐affinity state is preferentially recognized by agonists in vitro. For this reason, agonist tracers have been developed as tools for the noninvasive imaging of the high‐affinity state with positron emission tomography (PET). This review provides an overview of agonist tracers that have been developed for PET imaging of the brain, and the experimental paradigms that have been developed for the estimation of the relative abundance of receptors configured in the high‐affinity state. Agonist tracers appear to be more sensitive to endogenous neurotransmitter challenge than antagonists, as was originally expected. However, other expectations regarding agonist tracers have not been fulfilled. Potential reasons for difficulties in detecting the high‐affinity state in vivo are discussed.
Collapse
Affiliation(s)
- Vladimir Shalgunov
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine, Ghent University, University Hospital, Ghent, Belgium
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
11
|
In vivo biased agonism at 5-HT 1A receptors: characterisation by simultaneous PET/MR imaging. Neuropsychopharmacology 2018; 43:2310-2319. [PMID: 30030540 PMCID: PMC6135772 DOI: 10.1038/s41386-018-0145-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/07/2018] [Accepted: 06/27/2018] [Indexed: 12/26/2022]
Abstract
In neuropharmacology, the recent concept of 'biased agonism' denotes the capacity of certain agonists to target-specific intracellular pathways of a given receptor in specific brain areas. In the context of serotonin pharmacotherapy, 5-HT1A receptor-biased agonists could be of great interest in several neuropsychiatric disorders. The aim of this study was to determine whether biased agonists could be differentiated in terms of regional targeting by use of simultaneous functional magnetic resonance imaging (fMRI) and positron emission tomography (PET) brain imaging. We compared two 5-HT1A-biased agonists, NLX-112 and NLX-101, injected at three different doses in anaesthetised cats (n = 4). PET imaging was acquired for 90 min after bolus administration followed by constant infusion of the 5-HT1A radiotracer, [18F]MPPF. Drug occupancy was evaluated after injection at 50 min and BOLD fMRI was simultaneously acquired to evaluate subsequent brain activation patterns. 5-HT1A receptor occupancy was found to be dose-dependent for both agonists, but differed in magnitude and spatial distribution at equal doses with distinct BOLD patterns. Functional connectivity, as measured by BOLD signal temporal correlations between regions, was also differently modified by NLX-112 or NLX-101. Voxel-based correlation analyses between PET and fMRI suggested that NLX-112 stimulates both 5-HT1A autoreceptors and post-synaptic receptors, whereas NLX-101 preferentially stimulates post-synaptic cortical receptors. In cingulate cortex, the agonists induced opposite BOLD signal changes in response to receptor occupancy. These data constitute the first simultaneous exploration of 5-HT1A occupancy and its consequences in terms of brain activation, and demonstrates differential signalling by two 5-HT1A-biased agonists. Combined PET/fMRI represents a powerful tool in neuropharmacology, and opens new ways to address the concept of biased agonism by translational approaches.
Collapse
|
12
|
18F-F13640 preclinical evaluation in rodent, cat and primate as a 5-HT 1A receptor agonist for PET neuroimaging. Brain Struct Funct 2018; 223:2973-2988. [PMID: 29730825 DOI: 10.1007/s00429-018-1672-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/20/2018] [Indexed: 12/23/2022]
Abstract
Serotonin 1A receptors are known to play an important role in many psychiatric and neurodegenerative disorders. Currently, all available 5-HT1A receptor PET radiopharmaceuticals that are radiolabeled with fluorine-18 are antagonists. As agonists bind preferentially to the high-affinity state of receptors, it would be of great interest to develop agonist radioligands which could provide a measure of the functional 5-HT1A receptors in pathophysiological processes. The 5-HT1A receptor agonist candidates we recently proposed had promising in vitro properties but were not optimal in terms of PET imaging. F13640, a.k.a befiradol or NLX-112, is a 5-HT1A receptor agonist with a high affinity (Ki = 1 nM) and a high selectivity that would be suitable for a potential PET radiopharmaceutical. With propose here the first preclinical evaluation of 18F-F13640. 18F-F13640's nitro-precursor was synthesized and radiolabeled via a fluoro-nucleophilic substitution. Its radiopharmacological characterization included autoradiographic studies, metabolic studies, and in vivo PET scans in rat, cat and non-human primate. Some of the results were compared with the radiotracer 18F-MPPF, a 5-HT1A receptor antagonist. The radiochemical purity of 18F-F13640 was > 98%. In vitro binding pattern was consistent with the 5-HT1A receptor distribution. Metabolic studies revealed that the radiotracer rapidly entered the brain and led to few brain radiometabolites. Although 18F-F13640 in vivo binding was blocked by the 5-HT1A antagonist WAY-100635 and the 5-HT1A agonist 8-OH-DPAT, the distribution pattern was markedly different from antagonist radiotracers in the three species, suggesting it provides novel information on 5-HT1A receptors. Preliminary studies also suggest a high sensitivity of 18F-F13640 to endogenous serotonin release. 18F-F13640 has suitable characteristics for probing in vitro and in vivo the 5-HT1A receptors in high-affinity state. Quantification analyses with kinetic modeling are in progress to prepare the first-in-man study of 18F-F13640.
Collapse
|
13
|
Selvaraj S, Walker C, Arnone D, Cao B, Faulkner P, Cowen PJ, Roiser JP, Howes O. Effect of Citalopram on Emotion Processing in Humans: A Combined 5-HT 1A [ 11C]CUMI-101 PET and Functional MRI Study. Neuropsychopharmacology 2018; 43:655-664. [PMID: 28776580 PMCID: PMC5693328 DOI: 10.1038/npp.2017.166] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/18/2017] [Accepted: 08/01/2017] [Indexed: 12/21/2022]
Abstract
A subset of patients started on a selective serotonin reuptake inhibitor (SSRI) initially experience increased anxiety, which can lead to early discontinuation before therapeutic effects are manifest. The neural basis of this early SSRI effect is not known. Presynaptic dorsal raphe neuron (DRN) 5-HT1A receptors are known to have a critical role in affect processing. Thus we investigated the effect of acute citalopram on emotional processing and the relationship between DRN 5-HT1A receptor availability and amygdala reactivity. Thirteen (mean age 48±9 years) healthy male subjects received either a saline or citalopram infusion intravenously (10 mg over 30 min) on separate occasions in a single-blind, random order, crossover design. On each occasion, participants underwent a block design face-emotion processing task during fMRI known to activate the amygdala. Ten subjects also completed a positron emission tomography (PET) scan to quantify DRN 5-HT1A availability using [11C]CUMI-101. Citalopram infusion when compared with saline resulted in a significantly increased bilateral amygdala responses to fearful vs neutral faces (left p=0.025; right p=0.038 FWE-corrected). DRN [11C]CUMI-101 availability significantly positively correlated with the effect of citalopram on the left amygdala response to fearful faces (Z=2.51, p=0.027) and right amygdala response to happy faces (Z=2.33, p=0.032). Our findings indicate that the initial effect of SSRI treatment is to alter processing of aversive stimuli and that this is linked to DRN 5-HT1A receptors in line with evidence that 5-HT1A receptors have a role in mediating emotional processing.
Collapse
Affiliation(s)
- Sudhakar Selvaraj
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA,Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, London, UK,Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Biomedical and Behavioral Sciences Building (BBSB), 1941 East Road, Suite 3208 Houston, TX 77054, USA, Tel: +1 713 486 2500, Fax: +1 713 486 2553, E-mail:
| | - Chris Walker
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Danilo Arnone
- Institute of Psychiatry, King’s College London, Centre for Affective Disorders, London, UK,IoPPN, King’s College London, Institute of Psychiatry, Psychosis Studies, London, UK
| | - Bo Cao
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul Faulkner
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Philip J Cowen
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Jonathan P Roiser
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Oliver Howes
- Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, London, UK,IoPPN, King’s College London, Institute of Psychiatry, Psychosis Studies, London, UK,Institute of Clinical Sciences, Imperial College, Hammersmith Hospital, London, UK
| |
Collapse
|
14
|
Hazari PP, Pandey A, Chaturvedi S, Mishra AK. New Trends and Current Status of Positron-Emission Tomography and Single-Photon-Emission Computerized Tomography Radioligands for Neuronal Serotonin Receptors and Serotonin Transporter. Bioconjug Chem 2017; 28:2647-2672. [PMID: 28767225 DOI: 10.1021/acs.bioconjchem.7b00243] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The critical role of serotonin (5-hydroxytryptamine; 5-HT) and its receptors (5-HTRs) in the pathophysiology of diverse neuropsychiatric and neurodegenerative disorders render them attractive diagnostic and therapeutic targets for brain disorders. Therefore, the in vivo assessment of binding of 5-HT receptor ligands under a multitude of physiologic and pathologic scenarios may support more-accurate identification of disease and its progression and the patient's response to therapy as well as the screening of novel therapeutic strategies. The present Review aims to focus on the current status of radioligands used for positron-emission tomography (PET) and single-photon-emission computerized tomography (SPECT) imaging of human brain serotonin receptors. We further elaborate upon and emphasize the attributes that qualify a radioligand for theranostics on the basis of its frequency of use in clinics, its benefit to risk assessment in humans, and its continuous evolution, along with the major limitations.
Collapse
Affiliation(s)
- Puja Panwar Hazari
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences , Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Ankita Pandey
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences , Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Shubhra Chaturvedi
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences , Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Anil Kumar Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences , Brig S.K. Mazumdar Road, Delhi 110054, India
| |
Collapse
|
15
|
Edgar FG, Hansen HD, Leth-Petersen S, Ettrup A, Kristensen JL, Knudsen GM, Herth MM. Synthesis, radiofluorination, and preliminary evaluation of the potential 5-HT 2A receptor agonists [ 18 F]Cimbi-92 and [ 18 F]Cimbi-150. J Labelled Comp Radiopharm 2017; 60:586-591. [PMID: 28856700 DOI: 10.1002/jlcr.3557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/25/2022]
Abstract
An agonist PET tracer is of key interest for the imaging of the 5-HT2A receptor, as exemplified by the previously reported success of [11 C]Cimbi-36. Fluorine-18 holds several advantages over carbon-11, making it the radionuclide of choice for clinical purposes. In this respect, an 18 F-labelled agonist 5-HT2A receptor (5-HT2A R) tracer is highly sought after. Herein, we report a 2-step, 1-pot labelling methodology of 2 tracer candidates. Both ligands display high in vitro affinities for the 5-HT2A R. The compounds were synthesised from easily accessible labelling precursors, and radiolabelled in acceptable radiochemical yields, sufficient for in vivo studies in domestic pigs. PET images partially conformed to the expected brain distribution of the 5-HT2A R; a notable exception however being significant uptake in the striatum and thalamus. Additionally, a within-scan displacement challenge with a 5-HT2A R antagonist was unsuccessful, indicating that the tracers cannot be considered optimal for neuroimaging of the 5-HT2A R.
Collapse
Affiliation(s)
- Fraser Graeme Edgar
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Hanne D Hansen
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark
| | | | - Anders Ettrup
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark
| | - Jesper L Kristensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen, Denmark
| |
Collapse
|
16
|
Shrestha SS, Liow JS, Jenko K, Ikawa M, Zoghbi SS, Innis RB. The 5-HT1A Receptor PET Radioligand 11C-CUMI-101 Has Significant Binding to α1-Adrenoceptors in Human Cerebellum, Limiting Its Use as a Reference Region. J Nucl Med 2016; 57:1945-1948. [PMID: 27587705 DOI: 10.2967/jnumed.116.174151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/19/2016] [Indexed: 11/16/2022] Open
Abstract
Prazosin, a potent and selective α1-adrenoceptor antagonist, displaces 25% of 11C-CUMI-101 ([O-methyl-11C]2-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)dione) binding in monkey cerebellum. We sought to estimate the percentage contamination of 11C-CUMI-101 binding to α1-adrenoceptors in human cerebellum under in vivo conditions. In vitro receptor-binding techniques were used to measure α1-adrenoceptor density and the affinity of CUMI-101 for these receptors in human, monkey, and rat cerebellum. METHODS Binding potential (maximum number of binding sites × affinity [(1/dissociation constant]) was determined using in vitro homogenate binding assays in human, monkey, and rat cerebellum. 3H-prazosin was used to determine the maximum number of binding sites, as well as the dissociation constant of 3H-prazosin and the inhibition constant of CUMI-101. RESULTS α1-adrenoceptor density and the affinity of CUMI-101 for these receptors were similar across species. Cerebellar binding potentials were 3.7 for humans, 2.3 for monkeys, and 3.4 for rats. CONCLUSION Reasoning by analogy, 25% of 11C-CUMI-101 uptake in human cerebellum reflects binding to α1-adrenoceptors, suggesting that the cerebellum is of limited usefulness as a reference tissue for quantification in human studies.
Collapse
Affiliation(s)
- Stal S Shrestha
- Molecular Imaging Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Kimberly Jenko
- Molecular Imaging Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Masamichi Ikawa
- Molecular Imaging Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| |
Collapse
|
17
|
Kumar JSD, Underwood MD, Simpson NR, Kassir SA, Prabhakaran J, Majo VJ, Bakalian MJ, Parsey RV, Mann JJ, Arango V. Autoradiographic Evaluation of [(18)F]FECUMI-101, a High Affinity 5-HT1AR Ligand in Human Brain. ACS Med Chem Lett 2016; 7:482-6. [PMID: 27190597 DOI: 10.1021/acsmedchemlett.5b00499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/13/2016] [Indexed: 12/29/2022] Open
Abstract
[(18)F]FECUMI-101 ([(18)F]1) is a 5HT1AR ligand demonstrating specific binding in brain regions corresponding to the distribution of 5-HT1AR in baboons. However, we detected moderate uptake of [(18)F]1 in baboon thalamus, a brain region lacking 5-HT1AR. We sought to investigate the relative binding of [(18)F]1 to 5-HT1AR, α1R, and 5-HT7R in vitro. Using autoradiography in human brain sections, specific binding of [(18)F]1 to 5-HT1AR was confirmed. However, [(18)F]1 also showed 26% binding to α1R in PFC. The hippocampal formation exhibited 51% and 92% binding of [(18)F]1 to α1R and 5-HT1AR, respectively. Thalamus and cerebellum showed very little binding. There is no measurable specific binding of [(18)F]1 to 5-HT7R and no effect of temperature on [(18)F]1 specific binding to 5-HT1AR or α1R. These results indicate that, while [(18)F]FECUMI-101 is not a completely selective 5-HT1AR ligand for receptor quantification, it may be useful for occupancy measurements of drugs acting at 5-HT1AR in vivo.
Collapse
Affiliation(s)
- J. S. Dileep Kumar
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
- Department
of Psychiatry, Stony Brook University School of Medicine, Stony Brook, New York 11794, United States
| | - Mark D. Underwood
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
- Department
of Psychiatry, Columbia University Medical Center, New York, New York 10032, United States
| | - Norman R. Simpson
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Suham A. Kassir
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Jaya Prabhakaran
- Department
of Psychiatry, Columbia University Medical Center, New York, New York 10032, United States
| | - Vattoly J. Majo
- Department
of Psychiatry, Columbia University Medical Center, New York, New York 10032, United States
| | - Mihran J. Bakalian
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Ramin V. Parsey
- Department
of Psychiatry, Stony Brook University School of Medicine, Stony Brook, New York 11794, United States
| | - J. John Mann
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
- Department
of Psychiatry, Columbia University Medical Center, New York, New York 10032, United States
| | - Victoria Arango
- Division
of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York 10032, United States
- Department
of Psychiatry, Columbia University Medical Center, New York, New York 10032, United States
| |
Collapse
|
18
|
Pike VW. Considerations in the Development of Reversibly Binding PET Radioligands for Brain Imaging. Curr Med Chem 2016; 23:1818-69. [PMID: 27087244 PMCID: PMC5579844 DOI: 10.2174/0929867323666160418114826] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/04/2016] [Accepted: 04/15/2016] [Indexed: 12/17/2022]
Abstract
The development of reversibly binding radioligands for imaging brain proteins in vivo, such as enzymes, neurotransmitter transporters, receptors and ion channels, with positron emission tomography (PET) is keenly sought for biomedical studies of neuropsychiatric disorders and for drug discovery and development, but is recognized as being highly challenging at the medicinal chemistry level. This article aims to compile and discuss the main considerations to be taken into account by chemists embarking on programs of radioligand development for PET imaging of brain protein targets.
Collapse
Affiliation(s)
- Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Rm. B3C346A, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Kumar JSD, Mann JJ. PET tracers for serotonin receptors and their applications. Cent Nerv Syst Agents Med Chem 2015; 14:96-112. [PMID: 25360773 DOI: 10.2174/1871524914666141030124316] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/26/2014] [Accepted: 10/28/2014] [Indexed: 11/22/2022]
Abstract
Serotonin receptors (5-HTRs) are implicated in the pathophysiology of a variety of neuropsychiatric and neurodegenerative disorders and are also targets for drug therapy. In the CNS, most of these receptors are expressed in high abundance in specific brain regions reflecting their role in brain functions. Quantifying binding to 5-HTRs in vivo may permit assessment of physiologic and pathologic conditions, and monitoring disease progression, evaluating treatment response, and for investigating new treatment modalities. Positron emission tomography (PET) molecular imaging has the sensitivity to quantify binding of 5-HTRs in CNS disorders and to measure drug occupancy as part of a process of new drug development. Although research on PET imaging of 5-HTRs have been performed more than two decades, the successful radiotracers so far developed for human studies are limited to 5-HT₁AR, 5-HT₁BR, 5-HT₂AR, 5-HT₄R and 5-HT₆R. Herein we review the development and application of radioligands for PET imaging of 5-HTRs in living brain.
Collapse
Affiliation(s)
| | - J John Mann
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric institute, 1051 Riverside Drive, Box: 42, New York, NY, 10032, USA.
| |
Collapse
|
20
|
Optimising PET approaches to measuring 5-HT release in human brain. Synapse 2015; 69:505-11. [DOI: 10.1002/syn.21835] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 05/28/2015] [Accepted: 06/08/2015] [Indexed: 01/16/2023]
|
21
|
Finnema SJ, Scheinin M, Shahid M, Lehto J, Borroni E, Bang-Andersen B, Sallinen J, Wong E, Farde L, Halldin C, Grimwood S. Application of cross-species PET imaging to assess neurotransmitter release in brain. Psychopharmacology (Berl) 2015; 232:4129-57. [PMID: 25921033 PMCID: PMC4600473 DOI: 10.1007/s00213-015-3938-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/09/2015] [Indexed: 01/03/2023]
Abstract
RATIONALE This review attempts to summarize the current status in relation to the use of positron emission tomography (PET) imaging in the assessment of synaptic concentrations of endogenous mediators in the living brain. OBJECTIVES Although PET radioligands are now available for more than 40 CNS targets, at the initiation of the Innovative Medicines Initiative (IMI) "Novel Methods leading to New Medications in Depression and Schizophrenia" (NEWMEDS) in 2009, PET radioligands sensitive to an endogenous neurotransmitter were only validated for dopamine. NEWMEDS work-package 5, "Cross-species and neurochemical imaging (PET) methods for drug discovery", commenced with a focus on developing methods enabling assessment of changes in extracellular concentrations of serotonin and noradrenaline in the brain. RESULTS Sharing the workload across institutions, we utilized in vitro techniques with cells and tissues, in vivo receptor binding and microdialysis techniques in rodents, and in vivo PET imaging in non-human primates and humans. Here, we discuss these efforts and review other recently published reports on the use of radioligands to assess changes in endogenous levels of dopamine, serotonin, noradrenaline, γ-aminobutyric acid, glutamate, acetylcholine, and opioid peptides. The emphasis is on assessment of the availability of appropriate translational tools (PET radioligands, pharmacological challenge agents) and on studies in non-human primates and human subjects, as well as current challenges and future directions. CONCLUSIONS PET imaging directed at investigating changes in endogenous neurochemicals, including the work done in NEWMEDS, have highlighted an opportunity to further extend the capability and application of this technology in drug development.
Collapse
Affiliation(s)
- Sjoerd J. Finnema
- />Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Mika Scheinin
- />Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland , />Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Mohammed Shahid
- />Research and Development, Orion Corporation, Orion Pharma, Turku, Finland
| | - Jussi Lehto
- />Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland
| | - Edilio Borroni
- />Neuroscience Department, Hoffman-La Roche, Basel, Switzerland
| | | | - Jukka Sallinen
- />Research and Development, Orion Corporation, Orion Pharma, Turku, Finland
| | - Erik Wong
- />Neuroscience Innovative Medicine Unit, AstraZeneca, Wilmington, DE USA
| | - Lars Farde
- />Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden , />Translational Science Center at Karolinska Institutet, AstraZeneca, Stockholm, Sweden
| | - Christer Halldin
- />Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Sarah Grimwood
- Neuroscience Research Unit, Pfizer Inc, Cambridge, MA, USA. .,, 610 Main Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
22
|
Noninvasive blood-free full quantification of positron emission tomography radioligand binding. J Cereb Blood Flow Metab 2015; 35:148-56. [PMID: 25370860 PMCID: PMC4294408 DOI: 10.1038/jcbfm.2014.191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/19/2014] [Accepted: 10/08/2014] [Indexed: 11/09/2022]
Abstract
Full quantification of a positron emission tomography (PET) radioligand binding to its target is preferred because it requires the fewest assumptions, but generally involves measuring the concentration of free radioligand in the arterial plasma by collecting blood samples from the subject's radial artery during the scan, and performing metabolite analysis. This invasive, costly procedure deters subjects' participation, and requires specialized staff and equipment. Simultaneous estimation (SIME) can fully quantify binding using only PET data from multiple brain regions and one individual anchor value, which is based on a single arterial blood sample. Drawing this sample can still be challenging in clinical settings, particularly when using simultaneous PET/magnetic resonance scanners. Here we propose a methodology for full quantification of binding that does not require any blood samples. The methodology substitutes the SIME blood-based anchor with a value predicted using multiple linear regression of noninvasive, easy-to-collect variables related to the radioligand blood concentration, and individual metabolism, such as injected dose, body mass index, or body surface area. As a study case, we show here the methodology in comparison to analysis with full arterial-line blood sampling in a cohort of 23 available scans with [(11)C]CUMI-101, a partial agonist of the serotonin 5-HT1A receptors.
Collapse
|
23
|
Kumar JSD, Majo VJ, Prabhakaran J, Mann JJ. Synthesis and evaluation of arylpiperazines derivatives of 3,5-dioxo-(2H,4H)-1,2,4-triazine as 5-HT1AR ligands. Bioorg Med Chem Lett 2014; 24:4759-4762. [PMID: 25182564 DOI: 10.1016/j.bmcl.2014.07.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 07/16/2014] [Accepted: 07/17/2014] [Indexed: 12/23/2022]
Abstract
5-HT1AR agonist or partial agonists are established drug candidates for psychiatric and neurological disorders. We have reported the synthesis and evaluation of a series of high affinity 5-HT1AR partial agonist PET imaging agents with greater selectivity over α-1AR. The characteristic of these molecules are 3,5-dioxo-(2H,4H)-1,2,4-triazine skeleton tethered to an arylpiperazine unit through an alkyl side chain. The most potent 5-HT1AR agonistic properties were found to be associated with the molecules bearing C-4 alkyl group as the linker. Therefore development of 3,5-dioxo-(2H,4H)-1,2,4-triazine bearing arylpiperazine derivatives may provide high affinity selective 5-HT1AR ligands. Herein we describe the synthesis and evaluation of the binding properties of a series of arylpiperazine analogues of 3,5-dioxo-(2H,4H)-1,2,4-triazine.
Collapse
Affiliation(s)
- J S Dileep Kumar
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, USA; Department of Psychiatry and Behavior Science, Stony Brook University, New York, USA.
| | - Vattoly J Majo
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, USA
| | - Jaya Prabhakaran
- Columbia University College of Physicians and Surgeons, New York, USA
| | - J John Mann
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, USA; Columbia University College of Physicians and Surgeons, New York, USA
| |
Collapse
|
24
|
Shrestha SS, Liow JS, Lu S, Jenko K, Gladding RL, Svenningsson P, Morse CL, Zoghbi SS, Pike VW, Innis RB. (11)C-CUMI-101, a PET radioligand, behaves as a serotonin 1A receptor antagonist and also binds to α(1) adrenoceptors in brain. J Nucl Med 2014; 55:141-6. [PMID: 24385311 DOI: 10.2967/jnumed.113.125831] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The PET radioligand (11)C-CUMI-101 was previously suggested as a putative agonist radioligand for the serotonin 1A (5-hydroxytryptamine 1A [5-HT1A]) receptor in recombinant cells expressing human 5-HT1A receptor. However, a recent study showed that CUMI-101 behaved as a potent 5-HT1A receptor antagonist in rat brain. CUMI-101 also has moderate affinity (Ki = 6.75 nM) for α1 adrenoceptors measured in vitro. The current study examined the functional properties and selectivity of CUMI-101, both in vitro and in vivo. METHODS The functional assay was performed using (35)S-GTPγS (GTP is guanosine triphosphate) in primate brains. The cross-reactivity of CUMI-101 with α1 adrenoceptors was performed using in vitro radioligand binding studies in rat, monkey, and human brains as well as in vivo PET imaging in mouse, rat, and monkey brains. RESULTS CUMI-101 did not stimulate (35)S-GTPγS binding in primate brain, in contrast to 8-OH-DPAT, a potent 5-HT1A receptor agonist. Instead, CUMI-101 behaved as a potent 5-HT1A receptor antagonist by dose-dependently inhibiting 8-OH-DPAT-stimulated (35)S-GTPγS binding. Both in vitro and in vivo studies showed that CUMI-101 had significant α1 adrenoceptor cross-reactivity. On average, across all 3 species examined, cross-reactivity was highest in the thalamus (>45%) and lowest in the neocortex and cerebellum (<10%). PET imaging further confirmed that only preblocking with WAY-100635 plus prazosin decreased (11)C-CUMI-101 brain uptake to that of self-block. CONCLUSION CUMI-101 behaves as a 5-HT1A receptor antagonist in primate brain, with significant, regional-dependent α1 adrenoceptor cross-reactivity, limiting its potential use as a PET radioligand in humans.
Collapse
Affiliation(s)
- Stal Saurav Shrestha
- National Institute of Mental Health Intramural Research Program, Bethesda, Maryland; and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Majo VJ, Milak MS, Prabhakaran J, Mali P, Savenkova L, Simpson NR, Mann JJ, Parsey RV, Dileep Kumar JS. Synthesis and in vivo evaluation of [(18)F]2-(4-(4-(2-(2-fluoroethoxy)phenyl)piperazin-1-yl)butyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)-dione ([(18)F]FECUMI-101) as an imaging probe for 5-HT1A receptor agonist in nonhuman primates. Bioorg Med Chem 2013; 21:5598-604. [PMID: 23816046 PMCID: PMC3858174 DOI: 10.1016/j.bmc.2013.05.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/09/2013] [Accepted: 05/17/2013] [Indexed: 10/26/2022]
Abstract
The 5-HT1AR partial agonist PET radiotracer, [(11)C]CUMI-101, has advantages over an antagonist radiotracer as it binds preferentially to the high affinity state of the receptor and thereby provides more functionally meaningful information. The major drawback of C-11 tracers is the lack of cyclotron facility in many health care centers thereby limiting widespread clinical or research use. We identified the fluoroethyl derivative, 2-(4-(4-(2-(2-fluoroethoxy)phenyl)piperazin-1-yl)butyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)dione (FECUMI-101) (Ki=0.1nM; Emax=77%; EC50=0.65nM) as a partial agonist 5-HT1AR ligand of the parent ligand CUMI-101. FECUMI-101 is radiolabeled with F-18 by O-fluoroethylation of the corresponding desmethyl analogue (1) with [(18)F]fluoroethyltosylate in DMSO in the presence of 1.6equiv of K2CO3 in 45±5% yield (EOS). PET shows [(18)F]FECUMI-101 binds specifically to 5-HT1AR enriched brain regions of baboon. The specificity of [(18)F]FECUMI-101 binding to 5-HT1AR was confirmed by challenge studies with the known 5-HT1AR ligand WAY100635. These findings indicate that [(18)F]FECUMI-101 can be a viable agonist ligand for the in vivo quantification of high affinity 5-HT1AR with PET.
Collapse
Affiliation(s)
- Vattoly J. Majo
- Department of Psychiatry, Columbia University Medical Center, Columbia University, New York
| | - Matthew S. Milak
- Department of Psychiatry, Columbia University Medical Center, Columbia University, New York
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York
| | - Jaya Prabhakaran
- Department of Psychiatry, Columbia University Medical Center, Columbia University, New York
| | - Pratap Mali
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York
| | - Lyudmila Savenkova
- Department of Psychiatry, Columbia University Medical Center, Columbia University, New York
| | - Norman R. Simpson
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York
| | - J. John Mann
- Department of Psychiatry, Columbia University Medical Center, Columbia University, New York
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York
- Department of Radiology, Columbia University, New York, NY 10032
| | - Ramin V. Parsey
- Department of Psychiatry and Behavioral Science, Stony Brook University, New York
| | - J. S. Dileep Kumar
- Department of Psychiatry, Columbia University Medical Center, Columbia University, New York
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York
| |
Collapse
|
26
|
Zimmer L, Le Bars D. Current status of positron emission tomography radiotracers for serotonin receptors in humans. J Labelled Comp Radiopharm 2013; 56:105-13. [DOI: 10.1002/jlcr.3001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 11/08/2012] [Accepted: 11/08/2012] [Indexed: 12/19/2022]
|
27
|
Kumar JSD, Parsey RV, Kassir SA, Majo VJ, Milak MS, Prabhakaran J, Simpson NR, Underwood MD, Mann JJ, Arango V. Autoradiographic evaluation of [3H]CUMI-101, a novel, selective 5-HT1AR ligand in human and baboon brain. Brain Res 2013; 1507:11-8. [PMID: 23454434 DOI: 10.1016/j.brainres.2013.02.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 02/14/2013] [Accepted: 02/20/2013] [Indexed: 01/01/2023]
Abstract
[11C]CUMI-101 is the first selective serotonin receptor (5-HT1AR) partial agonist radiotracer for positron emission tomography (PET) tested in vivo in nonhuman primates and humans. We evaluated specific binding of [3H]CUMI-101 by quantitative autoradiography studies in postmortem baboon and human brain sections using the 5-HT1AR antagonist WAY-100635 as a displacer. The regional and laminar distributions of [3H]CUMI-101 binding in baboon and human brain sections matched the known distribution of [3H]8-OH-DPAT and [3H]WAY-100635. Prazosin did not measurably displace [3H]CUMI-101 binding in baboon or human brain sections, thereby ruling out [3H]CUMI-101 binding to α1-adrenergic receptors. This study demonstrates that [11C]CUMI-101 is a selective 5-HT1AR ligand for in vivo and in vitro studies in baboon and human brain.
Collapse
Affiliation(s)
- J S Dileep Kumar
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Paterson LM, Kornum BR, Nutt DJ, Pike VW, Knudsen GM. 5-HT radioligands for human brain imaging with PET and SPECT. Med Res Rev 2013; 33:54-111. [PMID: 21674551 PMCID: PMC4188513 DOI: 10.1002/med.20245] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The serotonergic system plays a key modulatory role in the brain and is the target for many drug treatments for brain disorders either through reuptake blockade or via interactions at the 14 subtypes of 5-HT receptors. This review provides the history and current status of radioligands used for positron emission tomography (PET) and single photon emission computerized tomography (SPECT) imaging of human brain serotonin (5-HT) receptors, the 5-HT transporter (SERT), and 5-HT synthesis rate. Currently available radioligands for in vivo brain imaging of the 5-HT system in humans include antagonists for the 5-HT(1A), 5-HT(1B), 5-HT(2A), and 5-HT(4) receptors, and for SERT. Here we describe the evolution of these radioligands, along with the attempts made to develop radioligands for additional serotonergic targets. We describe the properties needed for a radioligand to become successful and the main caveats. The success of a PET or SPECT radioligand can ultimately be assessed by its frequency of use, its utility in humans, and the number of research sites using it relative to its invention date, and so these aspects are also covered. In conclusion, the development of PET and SPECT radioligands to image serotonergic targets is of high interest, and successful evaluation in humans is leading to invaluable insight into normal and abnormal brain function, emphasizing the need for continued development of both SPECT and PET radioligands for human brain imaging.
Collapse
Affiliation(s)
- Louise M. Paterson
- Neuropsychopharmacology Unit, Division of Experimental Medicine, Imperial College London, Burlington Danes Building, Du Cane Road, London, United Kingdom
| | - Birgitte R. Kornum
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - David J. Nutt
- Neuropsychopharmacology Unit, Division of Experimental Medicine, Imperial College London, Burlington Danes Building, Du Cane Road, London, United Kingdom
| | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Gitte M. Knudsen
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
29
|
Measuring endogenous changes in serotonergic neurotransmission in humans: a [11C]CUMI-101 PET challenge study. Mol Psychiatry 2012; 17:1254-60. [PMID: 22665264 DOI: 10.1038/mp.2012.78] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Serotonin (5-HT) neurotransmission is implicated in cognitive and emotional processes and a number of neuropsychiatric disorders. The use of positron emission tomography (PET) to measure ligand displacement has allowed estimation of endogenous dopamine release in the human brain; however, applying this methodology to assess central 5-HT release has proved more challenging. The aim of this study was to assess the sensitivity of a highly selective 5-HT(1A) partial agonist radioligand [(11)C]CUMI-101 to changes in endogenous 5-HT levels induced by an intravenous challenge with the selective 5-HT re-uptake inhibitor (SSRI), citalopram, in healthy human participants. We studied 15 healthy participants who underwent PET scanning in conjunction with [(11)C]CUMI-101 after receiving an intravenous infusion of citalopram 10 mg or placebo in a double-blind, crossover, randomized design. Regional estimates of binding potential (BP(ND)) were obtained by calculating total volumes of distribution (V(T)) for presynaptic dorsal raphe nucleus (DRN) and postsynaptic cortical regions. Relative to placebo, citalopram infusion significantly increased [(11)C]CUMI-101 BP(ND) at postsynaptic 5-HT(1A) receptors in several cortical regions, but there was no change in binding at 5-HT(1A) autoreceptors in the DRN. Across the postsynaptic brain regions, citalopram treatment induced a mean 7% in [(11)C]CUMI-101 BP(ND) (placebo 1.3 (0.2); citalopram 1.4 (0.2); paired t-test P=0.003). The observed increase in postsynaptic [(11)C]CUMI-101 availability identified following acute citalopram administration could be attributable to a decrease in endogenous 5-HT availability in cortical terminal regions, consistent with preclinical animal studies, in which acute administration of SSRIs decreases DRN cell firing through activation of 5-HT(1A) autoreceptors to reduce 5-HT levels in postsynaptic regions. We conclude that [(11)C]CUMI-101 may be sensitive to changes in endogenous 5-HT release in humans.
Collapse
|
30
|
Kumar JSD, Milak MS, Majo VJ, Prabhakaran J, Mali P, Savenkova L, Mann JJ, Parsey RV. Comparison of high and low affinity serotonin 1A receptors by PET in vivo in nonhuman primates. J Pharmacol Sci 2012; 120:254-7. [PMID: 23076129 DOI: 10.1254/jphs.12100sc] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Serotonin (5-HT) 1A receptors exist in high and low affinity states. Agonist ligands bind preferentially to the high affinity state receptors, providing a more functionally relevant measure than antagonist binding. We now report comparison of 5-HT(1A) binding in vivo using both [¹¹C]CUMI-101 (agonist) and [¹¹C]WAY100635 (antagonist) in nonhuman primates. PET studies show that both tracers bind to known 5-HT(1A) receptor (5-HT(1A)R)-rich regions of baboon brain. The binding (BP(F)) of [¹¹C]CUMI-101 was lower on an average of 55% across the regions of interest (ROIs) compared to [¹¹C]WAY100635. This ratio is consistent with the in vitro binding data of agonist and antagonist 5-HT(1A)R ligands previously reported.
Collapse
Affiliation(s)
- J S Dileep Kumar
- Division of Molecular Imaging and Neuropathology, Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Pinborg LH, Feng L, Haahr ME, Gillings N, Dyssegaard A, Madsen J, Svarer C, Yndgaard S, Kjaer TW, Parsey RV, Hansen HD, Ettrup A, Paulson OB, Knudsen GM. No change in [¹¹C]CUMI-101 binding to 5-HT(1A) receptors after intravenous citalopram in human. Synapse 2012; 66:880-4. [PMID: 22730164 DOI: 10.1002/syn.21579] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 06/05/2012] [Accepted: 06/13/2012] [Indexed: 11/12/2022]
Abstract
The main objective of this study was to determine the sensitivity of [¹¹C]CUMI-101 to citalopram challenge aiming at increasing extracellular 5-HT. CUMI-101 has agonistic properties in human embryonic kidney 293 cells transfected with human recombinant 5-HT(1A) receptors (Hendry et al. [2011] Nucl Med Biol 38:273-277; Kumar et al. [2006] J Med Chem 49:125-134) and has previously been demonstrated to be sensitive to bolus citalopram in monkeys (Milak et al. [2011] J Cereb Blood Flow Metab 31:243-249). We studied six healthy individuals. Two PET-scans were performed on the same day in each individual before and after constant infusion of citalopram (0.15 mg/kg). The imaging data were analyzed using two tissue compartment kinetic modeling with metabolite corrected arterial input and Simplified Reference Tissue Modeling using cerebellum as a reference region. There was no significant difference in regional distribution volume or non-displaceable binding potential values before and after citalopram infusion. The mean receptor occupancy was 0.03 (range -0.14 to 0.17). Our data imply that [¹¹C]CUMI-101 binding is not sensitive to citalopram infusion in humans.
Collapse
Affiliation(s)
- Lars H Pinborg
- Center for Integrated Molecular Brain Imaging, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zimmer L, Luxen A. PET radiotracers for molecular imaging in the brain: Past, present and future. Neuroimage 2012; 61:363-70. [DOI: 10.1016/j.neuroimage.2011.12.037] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 12/15/2011] [Indexed: 12/22/2022] Open
|
33
|
Lemoine L, Becker G, Vacher B, Billard T, Lancelot S, Newman-Tancredi A, Zimmer L. Radiosynthesis and Preclinical Evaluation of 18F-F13714 as a Fluorinated 5-HT1A Receptor Agonist Radioligand for PET Neuroimaging. J Nucl Med 2012; 53:969-76. [DOI: 10.2967/jnumed.111.101212] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
34
|
Zoghbi SS, Anderson KB, Jenko KJ, Luckenbaugh DA, Innis RB, Pike VW. On quantitative relationships between drug-like compound lipophilicity and plasma free fraction in monkey and human. J Pharm Sci 2011; 101:1028-39. [PMID: 22170327 DOI: 10.1002/jps.22822] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 10/26/2011] [Accepted: 10/28/2011] [Indexed: 12/25/2022]
Abstract
Drug interactions with plasma proteins influence their pharmacokinetics and pharmacodynamics. We aimed to test whether a strong quantitative relationship exists between plasma free fraction (f(P) ) and lipophilicity for low molecular weight nonacidic drug-like compounds. We measured the n-octanol-buffer distribution coefficients at pH 7.4 ((m) logD) of 18 diverse radiotracers (<470 Da) used for brain imaging with positron emission tomography in vivo. Lipophilicities were also computed as (c) logD with two software packages. The f(P) values for monkeys and humans were determined by ultrafiltration and transformed into m logD(pr/pl) values representing the log(10) of the within phase partition of the radiotracers between plasma proteins and remaining plasma. (m) logD(pr/pl) correlated strongly with (m) logD for human ((m) logD(pr/pl) = 0.733(m) logD-0.780, r(2) = 0.74) and monkey ((m) logD(pr/pl) = 0.780(m) logD-1.15, r(2) = 0.83), but less strongly with (c) logD. These relationships were significantly different between species (P = 0.006). Removal of eight fluorinated compounds from the datasets raised r(2) to 0.81 and 0.91 for humans and monkeys, respectively. For the tested compounds, we conclude that n-octanol-buffer (pH 7.4) distribution strongly models that between plasma proteins and remaining plasma and moreover that (m) logD accounts for over 74% of compound (m) logD(pr/pl) and is a strong determinant of f(P).
Collapse
Affiliation(s)
- Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Kumar DJS, Bai B, Ng HH, Mirsalis JC, Erlandsson K, Milak MS, Majo VJ, Prabhakaran J, Mann JJ, Parsey RV. Biodistribution, toxicology, and radiation dosimetry of 5-HT1A-receptor agonist positron emission tomography ligand [11C]CUMI-101. Int J Toxicol 2011; 30:611-8. [PMID: 21994241 DOI: 10.1177/1091581811419024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sprague Dawley rats (10/sex/group) were given a single intravenous (iv) dose of CUMI-101 to determine acute toxicity of CUMI-101 and radiation dosimetry estimations were conducted in baboons with [(11)C]CUMI-101. Intravenous administration of CUMI-101 did not produce overt biologically or toxicologically significant adverse effects except transient hypoactivity immediately after dose in the mid- and high-dose groups, which is not considered to be a dose-limiting toxic effect. No adverse effects were observed in the low-dose group. The no observed adverse effect level (NOAEL) is considered to be 44.05 µg/kg for a single iv dose administration in rats. The maximum tolerated dose (MTD) was estimated to be 881 µg/kg for a single iv dose administration. The Medical Internal Radiation Dose (MIRDOSE) estimates indicate the maximum permissible single-study dosage of [(11)C]CUMI-101 in humans is 52 mCi with testes and urinary bladder as the critical organ for males and females, respectively.
Collapse
Affiliation(s)
- Dileep J S Kumar
- Department of Psychiatry and Division of Molecular Imaging and Neuropathology, Columbia University Medical Center and New York State Psychiatric Institute (NYSPI), New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hines CS, Liow JS, Zanotti-Fregonara P, Hirvonen J, Morse C, Pike VW, Innis RB. Human biodistribution and dosimetry of ¹¹C-CUMI-101, an agonist radioligand for serotonin-1a receptors in brain. PLoS One 2011; 6:e25309. [PMID: 21980419 PMCID: PMC3181260 DOI: 10.1371/journal.pone.0025309] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 08/31/2011] [Indexed: 12/04/2022] Open
Abstract
As a reported agonist,11C-CUMI-101 is believed to selectively bind the G-protein-coupled state of the serotonin-1A (5-HT1A) receptor, thereby providing a measure of the active subset of all 5-HT1A receptors in brain. Although 11C-CUMI-101 has been successfully used to quantify 5-HT1A receptors in human and monkey brain, its radiation exposure has not previously been reported. The purpose of this study was to calculate the radiation exposure to organs of the body based on serial whole-body imaging with positron emission tomography (PET) in human subjects.
Collapse
Affiliation(s)
- Christina S. Hines
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Jussi Hirvonen
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Cheryl Morse
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Robert B. Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
37
|
Malek-Saied N, Aissi RE, Ladeira S, Benoist E. Synthesis and biological evaluation of a novel 99mTc-cyclopentadienyltricarbonyl technetium complex as a new potential brain perfusion imaging agent. Appl Organomet Chem 2011. [DOI: 10.1002/aoc.1827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
38
|
Frankle WG, Lombardo I, Kegeles LS, Slifstein M, Martin JH, Huang Y, Hwang DR, Reich E, Cangiano C, Gil R, Abi-Dargham A, Laruelle M. Measurement of the serotonin 1A receptor availability in patients with schizophrenia during treatment with the antipsychotic medication ziprasidone. J Psychopharmacol 2011; 25:734-43. [PMID: 21109614 DOI: 10.1177/0269881110388329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of this study was to compare 5-HT(1A) availability in vivo in individuals with schizophrenia before and during treatment with the atypical antipsychotic ziprasidone. Six individuals with schizophrenia underwent two PET scans with [(11)C]WAY 100635; the first while medication-free (baseline) and the second while taking the atypical antipsychotic ziprasidone (on-medication). Regional volumes of distribution (V(T), mL g(-1)) were derived using a two-tissue compartment kinetic model. Outcome measures included binding potential relative to the plasma (BP(P), mL g(-1)) and the binding potential relative to the nonspecific distribution volume (BP(ND), unitless). No significant differences were observed in regional BP(P) or BP(ND) with ziprasidone treatment. A significant correlation was noted between BP(P) measured in the orbitofrontal cortex during the on-medication condition and degree of improvement in negative symptoms with treatment (r = 0.96, p = 0.004). Consistent with the published literature of changes in 5-HT(1A) binding during treatment with 5-HT(1A) receptor agonists, this study did not detect a significant reduction in 5-HT(1A) binding with ziprasidone. The finding of a relationship between 5-HT(1A) binding and the degree of improvement in negative symptoms provides further support for the role of the 5-HT(1A) receptor in the pathophysiology and treatment of this symptom domain.
Collapse
Affiliation(s)
- W Gordon Frankle
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Palner M, Underwood MD, Kumar DJS, Arango V, Knudsen GM, John Mann J, Parsey RV. Ex vivo evaluation of the serotonin 1A receptor partial agonist [³H]CUMI-101 in awake rats. Synapse 2011; 65:715-23. [PMID: 21108237 DOI: 10.1002/syn.20888] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 11/11/2010] [Indexed: 11/11/2022]
Abstract
[³H]CUMI-101 is a 5-HT(1A) partial agonist, which has been evaluated for use as a positron emission tracer in baboon and humans. We sought to evaluate the properties of [³H]CUMI-101 ex vivo in awake rats and determine if [³H]CUMI-101 can measure changes in synaptic levels of serotonin after different challenge paradigms. [³H]CUMI-101 shows good uptake and good specific binding ratio (SBR) in frontal cortex 5.18 and in hippocampus 3.18. Binding was inhibited in a one-binding-site fashion by WAY100635 and unlabeled CUMI-101. The ex vivo B(max) of [³H]CUMI-101 in frontal cortex (98.7 fmol/mg) and hippocampus (131 fmol/kg) agree with the ex vivo B(max) of [³H]MPPF in frontal cortex (147.1 fmol/mg) and hippocampus (72.1 fmol/mg) and with in vitro values reported with 8-OH-DPAT. Challenges with citalopram, a selective serotonin reuptake inhibitor, fenfluramine, a serotonin releaser, and 4-chloro-DL-phenylalanine, a serotonin synthesis inhibitor, did not show any effect on the standardized uptake values (SUVs) in any region. Citalopram did alter SBR, but this was due to changes in cerebellar SUVs. Our results indicate that [³H]CUMI-101 is a good radioligand for imaging 5-HT(1A) high-density regions in rats; however, the results from pharmacological challenges remain inconclusive.
Collapse
Affiliation(s)
- Mikael Palner
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
In vivo serotonin-sensitive binding of [11C]CUMI-101: a serotonin 1A receptor agonist positron emission tomography radiotracer. J Cereb Blood Flow Metab 2011; 31:243-9. [PMID: 20571518 PMCID: PMC3049488 DOI: 10.1038/jcbfm.2010.83] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Positron emission tomography studies of 5-hydroxytryptamine (5-HT)(1A) receptors have hitherto been limited to antagonist radiotracers. Antagonists do not distinguish high/low-affinity conformations of G protein-coupled receptors and are less likely to be sensitive to intrasynaptic serotonin levels. We developed a novel 5-HT(1A) agonist radiotracer [(11)C]CUMI-101. This study evaluates the sensitivity of [(11)C]CUMI-101 binding to increases in intrasynaptic serotonin induced by intravenous citalopram and fenfluramine. Two Papio anubis were scanned, using [(11)C]CUMI-101 intravenous bolus of 4.5 ± 1.5 mCi. Binding potential (BP(F)=B(avail)/K(D)) was measured before (n=10) and 20 minutes after elevation of intrasynaptic serotonin by intravenous citalopram (2 mg/kg, n=3; 4 mg/kg, n=3) and fenfluramine (2.5 mg/kg, n=3) using a metabolite-corrected arterial input function. Occupancy was also estimated by the Lassen graphical approach. Both citalopram and fenfluramine effects were significant for BP(F) (P=0.031, P=0.049, respectively). The Lassen approach estimated 15.0, 30.4, and 23.7% average occupancy after citalopram 2 mg/kg, 4 mg/kg, and fenfluramine 2.5 mg/kg, respectively. [(11)C]CUMI-101 binding is sensitive to a large increase in intrasynaptic serotonin in response to robust pharmacological challenges. These modest changes in BP(F) may make it unlikely that this ligand will detect changes in intrasynaptic 5-HT under physiologic conditions; future work will focus on evaluating its utility in measuring the responsiveness of the 5-HT system to pharmacological challenges.
Collapse
|
41
|
Seridi A, Wolff M, Boulay A, Saffon N, Coulais Y, Picard C, Machura B, Benoist E. Rhenium(I) and technetium(I) complexes of a novel pyridyltriazole-based ligand containing an arylpiperazine pharmacophore: Synthesis, crystal structures, computational studies and radiochemistry. INORG CHEM COMMUN 2011. [DOI: 10.1016/j.inoche.2010.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Hendry N, Christie I, Rabiner EA, Laruelle M, Watson J. In vitro assessment of the agonist properties of the novel 5-HT1A receptor ligand, CUMI-101 (MMP), in rat brain tissue. Nucl Med Biol 2010; 38:273-7. [PMID: 21315283 DOI: 10.1016/j.nucmedbio.2010.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/31/2010] [Accepted: 08/11/2010] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Development of agonist positron emission tomography (PET) radioligands for the 5-HT neurotransmitter system is an important target to enable the understanding of human 5-HT function in vivo. [(11)C]CUMI-101, proposed as the first 5-HT(1A) receptor agonist PET ligand, has been reported to behave as a potent 5-HT(1A) agonist in a cellular system stably expressing human recombinant 5-HT(1A) receptors. In this study, we investigate the agonist properties of CUMI-101 in rat brain tissue. METHODS [(35)S]-GTPγS binding studies were used to determine receptor function in HEK (human embryonic kidney) 293 cells transfected with human recombinant 5-HT(1A) receptors and in rat cortex and rat hippocampal tissue, following administration of CUMI-101 and standard 5-HT1A antagonists (5-HT, 5-CT and 8-OH-DPAT). RESULTS CUMI-101 behaved as an agonist at human recombinant 5-HT(1A) receptors (pEC(50) 9.2). However, CUMI-101 did not show agonist activity in either rat cortex or hippocampus at concentrations up to 10 μM. In these tissues, CUMI-behaved as an antagonist with pK(B)s of 9.2 and 9.3, respectively. CONCLUSIONS Our studies demonstrate that as opposed to its behavior in human recombinant system, in rat brain tissue CUMI-101 behaves as a potent 5-HT(1A) receptor antagonist.
Collapse
Affiliation(s)
- Nicola Hendry
- Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline, Harlow, CM19 5AW Essex, UK
| | | | | | | | | |
Collapse
|
43
|
Milak MS, DeLorenzo C, Zanderigo F, Prabhakaran J, Kumar JSD, Majo VJ, Mann JJ, Parsey RV. In vivo quantification of human serotonin 1A receptor using 11C-CUMI-101, an agonist PET radiotracer. J Nucl Med 2010; 51:1892-900. [PMID: 21098796 DOI: 10.2967/jnumed.110.076257] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The serotonin (5-hydroxytryptamine, or 5-HT) type 1A receptor (5-HT(1A)R) is implicated in the pathophysiology of numerous neuropsychiatric disorders. We have published the initial evaluation and reproducibility in vivo of [O-methyl-(11)C]2-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)dione ((11)C-CUMI-101), a novel 5-HT(1A) agonist radiotracer, in Papio anubis. Here, we report the optimal modeling parameters of (11)C-CUMI-101 for human PET studies. METHODS PET scans were obtained for 7 adult human volunteers. (11)C-CUMI-101 was injected as an intravenous bolus, and emission data were collected for 120 min in 3-dimensional mode. We evaluated 10 different models using metabolite-corrected arterial input functions or reference region approaches and several outcome measures. RESULTS When using binding potential (BP(F) = B(avail)/K(D) [total available receptor concentration divided by the equilibrium dissociation constant]) as the outcome measure, the likelihood estimation in the graphical analysis (LEGA) model performed slightly better than the other methods evaluated at full scan duration. The average test-retest percentage difference was 9.90% ± 5.60%. When using BP(ND) (BP(ND) = f(nd) × B(avail)/K(D); BP(ND) equals the product of BP(F) and f(nd) [free fraction in the nondisplaceable compartment]), the simplified reference tissue method (SRTM) achieved the lowest percentage difference and smallest bias when compared with nondisplaceable binding potential obtained from LEGA using the metabolite-corrected plasma input function (r(2) = 0.99; slope = 0.92). The time-stability analysis indicates that a 120-min scan is sufficient for the stable estimation of outcome measures. Voxel results were comparable to region-of-interest-based analysis, with higher spatial resolution. CONCLUSION On the basis of its measurable and stable free fraction, high affinity and selectivity, good blood-brain barrier permeability, and plasma and brain kinetics, (11)C-CUMI-101 is suitable for the imaging of high-affinity 5-HT(1A) binding in humans.
Collapse
Affiliation(s)
- Matthew S Milak
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Measuring serotonin synthesis: from conventional methods to PET tracers and their (pre)clinical implications. Eur J Nucl Med Mol Imaging 2010; 38:576-91. [PMID: 21113591 PMCID: PMC3034914 DOI: 10.1007/s00259-010-1663-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 11/01/2010] [Indexed: 12/20/2022]
Abstract
The serotonergic system of the brain is complex, with an extensive innervation pattern covering all brain regions and endowed with at least 15 different receptors (each with their particular distribution patterns), specific reuptake mechanisms and synthetic processes. Many aspects of the functioning of the serotonergic system are still unclear, partially because of the difficulty of measuring physiological processes in the living brain. In this review we give an overview of the conventional methods of measuring serotonin synthesis and methods using positron emission tomography (PET) tracers, more specifically with respect to serotonergic function in affective disorders. Conventional methods are invasive and do not directly measure synthesis rates. Although they may give insight into turnover rates, a more direct measurement may be preferred. PET is a noninvasive technique which can trace metabolic processes, like serotonin synthesis. Tracers developed for this purpose are α-[11C]methyltryptophan ([11C]AMT) and 5-hydroxy-L-[β-11C]tryptophan ([11C]5-HTP). Both tracers have advantages and disadvantages. [11C]AMT can enter the kynurenine pathway under inflammatory conditions (and thus provide a false signal), but this tracer has been used in many studies leading to novel insights regarding antidepressant action. [11C]5-HTP is difficult to produce, but trapping of this compound may better represent serotonin synthesis. AMT and 5-HTP kinetics are differently affected by tryptophan depletion and changes of mood. This may indicate that both tracers are associated with different enzymatic processes. In conclusion, PET with radiolabelled substrates for the serotonergic pathway is the only direct way to detect changes of serotonin synthesis in the living brain.
Collapse
|
45
|
Prabhakaran J, Majo VJ, Milak MS, Mali P, Savenkova L, Mann JJ, Parsey RV, Kumar JD. Synthesis and in vivo evaluation of [11C]MPTQ: A potential PET tracer for alpha2A-adrenergic receptors. Bioorg Med Chem Lett 2010; 20:3654-7. [DOI: 10.1016/j.bmcl.2010.04.099] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 04/20/2010] [Accepted: 04/21/2010] [Indexed: 12/28/2022]
|
46
|
Dawson LA, Watson JM. Vilazodone: a 5-HT1A receptor agonist/serotonin transporter inhibitor for the treatment of affective disorders. CNS Neurosci Ther 2010; 15:107-17. [PMID: 19499624 DOI: 10.1111/j.1755-5949.2008.00067.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Vilazodone (EMD 68843; 5-{4-[4-(5-cyano-3-indolyl)-butyl]-1-piperazinyl}-benzofuran-2-carboxamide hydrochloride) is a combined serotonin specific reuptake inhibitor (SSRI) and 5-HT1A receptor partial agonist currently under clinical evaluation for the treatment of major depression. This molecule was designed based on the premise that negative feedback circuitry, mediated via 5-HT1 receptors, limits the acute SSRI-induced enhancements in serotonergic neurotransmission. If the hypothesis is correct, combination of SSRI with 5-HT1A partial agonism should temporally enhance the neuroplastic adaptation and subsequently hasten therapeutic efficacy compared to current treatments. Preclinical in vitro evaluation has confirmed vilazodone's primary pharmacological profile both in clonal and native systems, that is, serotonin reuptake blockade and 5-HT1A partial agonism. However, in vivo and in contrast to combination of 8-OH-DPAT and paroxetine, vilazodone selectively enhanced serotonergic output in the prefrontal cortex of rats. Behavioral evaluations, in the ultrasonic vocalization model of anxiety in rats, demonstrated anxiolytic efficacy. In the forced swim test (a putative model of depression), vilazodone also showed efficacy but at a single dose only. In man, vilazodone abolished REM sleep and demonstrated clinical antidepressant efficacy equivalent to an SSRI. Ongoing clinical evaluations will hopefully reveal whether the founding hypothesis was valid and if vilazodone will produce a more rapid onset of antidepressant efficacy.
Collapse
Affiliation(s)
- Lee A Dawson
- Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline, Harlow, Essex, UK
| | | |
Collapse
|
47
|
Lu S, Liow JS, Zoghbi SS, Hong J, Innis RB, Pike VW. Evaluation of [C]S14506 and [F]S14506 in rat and monkey as agonist PET radioligands for brain 5-HT(1A) receptors. Curr Radiopharm 2010; 3:9-18. [PMID: 20657759 PMCID: PMC2908029 DOI: 10.2174/1874471011003010009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In vitro and ex vivo measurements have shown that the binding of the selective high-affinity agonist, S14506 (1-[2-(4-fluorobenzoylamino)ethyl]-4-(7-methoxy-naphthyl)piperazine), to 5-HT(1A) receptors, is similar in affinity (K(d) = 0.79 nM) and extent (B(max)) to that of the antagonist, WAY 100635. We aimed to test whether S14506, labeled with a positron-emitter, might serve as a radioligand for imaging brain 5-HT(1A) receptors in vivo with positron emission tomography (PET). Here we evaluated [(11)C]S14506 and [(18)F]S14506 in rat and rhesus monkey in vivo. After intravenous administration of [(11)C]S14506 into rat, radioactivity entered brain, reaching 210% SUV at 2 min. Radioactivity uptake into brain was higher (~ 350% SUV) in rats pre-treated with the P-glycoprotein (P-gp) inhibitor, cyclosporin A. In rhesus monkey, peak brain uptake of radioactivity after administration of [(11)C]S14506 or [(18)F]S14506 was also moderate and for [(11)C]S14506 increased from ~ 170% SUV after 7 min, to 240% SUV in a monkey pre-treated with the P-gp inhibitor, tariquidar. The ratios of radioactivity in 5-HT(1A) receptor-rich regions, such as cingulate or hippocampus to that in receptor-poor cerebellum reached between 1.35 and 1.5 at 60 min for both [(11)C]S14506 and [(18)F]S14506. [(11)C]S14506 gave one major polar radiometabolite in monkey plasma, and [(18)F]S14506 gave three and two more polar radiometabolites in rat and monkey plasma, respectively. The rat radiometabolites of [(18)F]S14506 did not accumulate in brain. [(18)F]S14506 was not radiodefluorinated in monkey. Thus, despite high-affinity and lack of troublesome brain radiometabolites, both [(11)C]S14506 and [(18)F]S14506 were ineffective for imaging rat or monkey brain 5-HT(1A) receptors in vivo, even under P-gp inhibited conditions. Explanations for the failure of these radioligands are offered.
Collapse
Affiliation(s)
- Shuiyu Lu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda MD 20892-1003, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda MD 20892-1003, USA
| | - Sami S. Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda MD 20892-1003, USA
| | - Jinsoo Hong
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda MD 20892-1003, USA
| | - Robert B. Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda MD 20892-1003, USA
| | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Room B3C346, Bethesda MD 20892-1003, USA
| |
Collapse
|
48
|
Lemoine L, Verdurand M, Vacher B, Blanc E, Le Bars D, Newman-Tancredi A, Zimmer L. [18F]F15599, a novel 5-HT1A receptor agonist, as a radioligand for PET neuroimaging. Eur J Nucl Med Mol Imaging 2009; 37:594-605. [PMID: 19789870 DOI: 10.1007/s00259-009-1274-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Accepted: 08/24/2009] [Indexed: 11/30/2022]
Abstract
PURPOSE The serotonin-1A (5-HT(1A)) receptor is implicated in the pathophysiology of major neuropsychiatric disorders. Thus, the functional imaging of 5-HT(1A) receptors by positron emission tomography (PET) may contribute to the understanding of its role in those pathologies and their therapeutics. These receptors exist in high- and low-affinity states and it is proposed that agonists bind preferentially to the high-affinity state of the receptor and therefore could provide a measure of the functional 5-HT(1A) receptors. Since all clinical PET 5-HT(1A) radiopharmaceuticals are antagonists, it is of great interest to develop a( 18)F labelled agonist. METHODS F15599 (3-chloro-4-fluorophenyl-(4-fluoro-4{[(5-methyl-pyrimidin-2-ylmethyl)-amino]-methyl}-piperidin-1-yl)-methanone) is a novel ligand with high affinity and selectivity for 5-HT(1A) receptors and is currently tested as an antidepressant. In pharmacological tests in rat, it exhibits preferential agonist activity at post-synaptic 5-HT(1A) receptors in cortical brain regions. Here, its nitro-precursor was synthesised and radiolabelled via a fluoronucleophilic substitution. Radiopharmacological evaluations included in vitro and ex vivo autoradiography in rat brain and PET scans on rats and cats. Results were compared with simultaneous studies using [(18)F]MPPF, a validated 5-HT(1A) antagonist radiopharmaceutical. RESULTS The chemical and radiochemical purities of [(18)F]F15599 were >98%. In vitro [(18)F]F15599 binding was consistent with the known 5-HT(1A) receptors distribution (hippocampus, dorsal raphe nucleus, and notably cortical areas) and addition of Gpp(NH)p inhibited [(18)F]F15599 binding, consistent with a specific binding to G protein-coupled receptors. In vitro binding of [(18)F]F15599 was blocked by WAY100635 and 8-OH-DPAT, respectively, prototypical 5-HT(1A) antagonist and agonist. The ex vivo and in vivo studies demonstrated that the radiotracer readily entered the rat and the cat brain and generated few brain radioactive metabolites. Remarkably, in microPET studies, [(18)F]F15599 notably displayed a pattern of brain labelling that did not correlate with in vitro observations. Thus, in cat, the highest binding was observed in dorsal raphe and cingulate cortex with little binding in other cortical regions and none in hippocampus. In vivo binding was abolished by WAY100635, indicating specific labelling of 5-HT(1A) receptors. CONCLUSION [(18)F]F15599 is a radiofluorinated agonist presenting interesting characteristics for probing in vitro and in vivo the high-affinity states of the 5-HT(1A) receptors. Its differential labelling of 5-HT(1A) receptors in vitro and in vivo may result from its reported preferential interaction with receptors coupled to specific G-protein subtypes.
Collapse
Affiliation(s)
- Laëtitia Lemoine
- Laboratory of Neuropharmacology, Université de Lyon, EAC CNRS 5006, Lyon, France
| | | | | | | | | | | | | |
Collapse
|
49
|
Wang F, Wang X, Yang S, Liu J, Zhang X. N-[3-[4-(2-methoxyphenyl) piperaziny-1-yl]propyl]cyclam: synthesized as a potential 5-HT1Areceptor ligand and labelled with99mTc-nitrido core. J Labelled Comp Radiopharm 2008. [DOI: 10.1002/jlcr.1532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
50
|
Milak MS, Severance AJ, Ogden RT, Prabhakaran J, Kumar JSD, Majo VJ, Mann JJ, Parsey RV. Modeling considerations for 11C-CUMI-101, an agonist radiotracer for imaging serotonin 1A receptor in vivo with PET. J Nucl Med 2008; 49:587-96. [PMID: 18344443 PMCID: PMC3580231 DOI: 10.2967/jnumed.107.046540] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Several lines of evidence demonstrate involvement of serotonin 1A receptors (5-HT1ARs) in the pathophysiology of neuropsychiatric disorders such as depression, suicidal behavior, schizophrenia, and Alzheimer's disease. We recently published the synthesis and initial evaluation of [O-methyl-11C]2-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)-4-methyl-1,2,4-triazine-3,5(2H,4H)dione (11C-MMP), a 5-HT1AR agonist. Here we determine the optimal modeling parameters for 11C-MMP under its new name, 11C-CUMI-101, in Papio anubis. METHODS PET scans were performed on 2 adult male P. anubis; 166.5 MBq +/- 43.0 (4.50 +/- 1.16 mCi) of 11C-CUMI-101 were injected as an intravenous bolus, and emission data were collected for 120 min in 3-dimensional mode. We evaluated 4 different models (1- and 2-tissue compartment iterative and noniterative kinetic models, basis pursuit, and likelihood estimation in graphical analysis [LEGA]), using binding potential (BPF = Bmax/Kd) (Bmax = maximum number of binding sites; Kd = dissociation constant) as the outcome measure. Arterial blood sampling and metabolite-corrected arterial input function were used for full quantification of BPF. To assess the performance of each model, we compared results using 6 different metrics (percentage difference, within-subject mean sum of squares [WSMSS] for reproducibility; variance across subjects, intraclass correlation coefficient [ICC] for reliability; identifiability based on bootstrap resampling of residuals; and time stability analysis to determine minimal required scanning time) at each of 6 different scanning durations. Models were also evaluated on scans acquired after injecting the 5-HT1A antagonist [N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl) cyclohexane carboxamide] [WAY100635] 0.5 mg/kg, intravenous) and the 5-HT1A agonist 8-hydroxy-2-(di-n-propylamino)tetralin) [8-OH-DPAT] 2 mg/kg, intravenous). RESULTS All metabolites are more polar than 11C-CUMI-101, and no significant change in metabolites was observed in the blocking studies. The free fraction is 59% +/- 3%. We determined that 100 min of scanning time is adequate and that for the region-of-interest (ROI)-level analysis, the LEGA model gives the best results. The median test-retest percentage difference for BPF is 11.15% +/- 4.82% across all regions, WSMSS = 2.66, variance = 6.07, ICC = 0.43, and bootstrap identifiability = 0.59. Preadministration of WAY100635 and 8-OH-DPAT resulted in 87% and 76% average reductions in BPF values, respectively, across ROIs. CONCLUSION On the basis of the measurable free fraction, high affinity and selectivity, adequate blood-brain permeability, and favorable plasma and brain kinetics, 11C-CUMI-101 is an excellent candidate for imaging high-affinity 5-HT1ARs in humans.
Collapse
Affiliation(s)
- Matthew S Milak
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| | | | | | | | | | | | | | | |
Collapse
|