1
|
Everix L, Elvas F, Miranda Menchaca A, Khetarpal V, Liu L, Bard J, Staelens S, Bertoglio D. Preclinical validation and kinetic modelling of the SV2A PET ligand [ 18F]UCB-J in mice. J Cereb Blood Flow Metab 2025; 45:920-931. [PMID: 39628318 PMCID: PMC11615906 DOI: 10.1177/0271678x241304923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/19/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
Synaptic vesicle protein 2A (SV2A) is ubiquitously expressed in presynaptic terminals where it functions as a neurotransmission regulator protein. Synaptopathy has been reported during healthy ageing and in a variety of neurodegenerative diseases. Positron emission tomography (PET) imaging of SV2A can be used to evaluate synaptic density. The PET ligand [11C]UCB-J has high binding affinity and selectivity for SV2A but has a short physical half-life due to the 11C isotope. Here we report the characterization and validation of its 18F-labeled equivalent, [18F]UCB-J, in terms of specificity, reproducibility and stability in C57BL/6J mice. Plasma analysis revealed at least one polar radiometabolite. Kinetic modelling was performed using a population-based metabolite corrected image-derived input function (IDIF). [18F]UCB-J showed relatively fast kinetics and a reliable measure of the IDIF-based volume of distribution (VT(IDIF)). [18F]UCB-J specificity for SV2A was confirmed through a levetiracetam blocking assay (50 to 200 mg/kg). Reproducibility of the VT(IDIF) was determined through test-retest analysis, revealing significant correlation (r2 = 0.773, p < 0.0001). Time-stability analyses indicate a scan duration of 60 min to be sufficient to obtain a reliable VT(IDIF). In conclusion, [18F]UCB-J is a selective SV2A ligand with optimal kinetics in mice. Further investigation is warranted for (pre)clinical applicability of [18F]UCB-J in synaptopathies.
Collapse
Affiliation(s)
- Liesbeth Everix
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Filipe Elvas
- Molecular Imaging and Radiology (MIRA), Wilrijk, Belgium
| | | | - Vinod Khetarpal
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, USA
| | - Longbin Liu
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, USA
| | - Jonathan Bard
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc., Princeton, NJ, USA
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
2
|
Lu X, Ji B, Huang G, Ding H. Advances in synaptic PET imaging and intervention with synapse-targeted small-molecular drugs for dementia diagnosis and therapy. FUNDAMENTAL RESEARCH 2025; 5:63-71. [PMID: 40166112 PMCID: PMC11955051 DOI: 10.1016/j.fmre.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/02/2025] Open
Abstract
Dementia is characterized by synaptic and neuronal dysfunction in disease-specific brain regions. Repeated failure of dementia clinical trials with therapeutic drugs targeting abnormal protein aggregates has caused researchers to shift their focus to synaptic functions and increased the importance of clinically available imaging for synaptic density and the development of synapse-targeted intervention. Synaptic density imaging with positron emission tomography (PET) tracer enables non-invasive detection of synaptic loss and hence investigates the association with other neuropathological events exemplified by disease-specific abnormal protein accumulation. Many studies have reviewed the progress of synaptic density imaging; however, to our knowledge, there is no article yet that summarizes the research progress of multimodal imaging of synaptic density tracers combined with other dementia biomarkers. Moreover, synaptic function intervention for dementia therapy has not yet been summarized. In this review, first we detail the progress of synaptic density imaging including tracer development and preclinical/clinical application, followed by a discussion of multimodal imaging of synaptic density tracers combined with classic dementia biomarkers in the clinical research stage. Finally, we briefly summarize the synapse-targeted drugs for dementia therapy.
Collapse
Affiliation(s)
- Xiuhong Lu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- School of pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of pharmacy, Fudan University, Shanghai 201203, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Hong Ding
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
3
|
Asch RH, Abdallah CG, Carson RE, Esterlis I. Challenges and rewards of in vivo synaptic density imaging, and its application to the study of depression. Neuropsychopharmacology 2024; 50:153-163. [PMID: 39039139 PMCID: PMC11525584 DOI: 10.1038/s41386-024-01913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024]
Abstract
The development of novel radiotracers for Positron Emission Tomography (PET) imaging agents targeting the synaptic vesicle glycoprotein 2 A (SV2A), an integral glycoprotein present in the membrane of all synaptic vesicles throughout the central nervous system, provides a method for the in vivo quantification of synaptic density. This is of particular interest in neuropsychiatric disorders given that synaptic alterations appear to underlie disease progression and symptom severity. In this review, we briefly describe the development of these SV2A tracers and the evaluation of quantification methods. Next, we discuss application of SV2A PET imaging to the study of depression, including a review of our findings demonstrating lower SV2A synaptic density in people with significant depressive symptoms and the use of a ketamine drug challenge to examine synaptogenesis in vivo. We then highlight the importance of performing translational PET imaging in animal models in conjunction with clinical imaging. We consider the ongoing challenges, possible solutions, and present preliminary findings from our lab demonstrating the translational benefit and potential of in vivo SV2A imaging in animal models of chronic stress. Finally, we discuss methodological improvements and future directions for SV2A imaging, potentially in conjunction with other neural markers.
Collapse
Affiliation(s)
- Ruth H Asch
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Chadi G Abdallah
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT, USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT, USA.
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
4
|
Howes O, Marcinkowska J, Turkheimer FE, Carr R. Synaptic changes in psychiatric and neurological disorders: state-of-the art of in vivo imaging. Neuropsychopharmacology 2024; 50:164-183. [PMID: 39134769 PMCID: PMC11525650 DOI: 10.1038/s41386-024-01943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 11/01/2024]
Abstract
Synapses are implicated in many neuropsychiatric illnesses. Here, we provide an overview of in vivo techniques to index synaptic markers in patients. Several positron emission tomography (PET) tracers for synaptic vesicle glycoprotein 2 A (SV2A) show good reliability and selectivity. We review over 50 clinical studies including over 1700 participants, and compare findings in healthy ageing and across disorders, including addiction, schizophrenia, depression, posttraumatic stress disorder, and neurodegenerative disorders, including tauopathies, Huntington's disease and α-synucleinopathies. These show lower SV2A measures in cortical brain regions across most of these disorders relative to healthy volunteers, with the most well-replicated findings in tauopathies, whilst changes in Huntington's chorea, Parkinson's disease, corticobasal degeneration and progressive supranuclear palsy are predominantly subcortical. SV2A PET measures are correlated with functional connectivity across brain networks, and a number of other measures of brain function, including glucose metabolism. However, the majority of studies found no relationship between grey matter volume measured with magnetic resonance imaging and SV2A PET measures. Cognitive dysfunction, in domains including working memory and executive function, show replicated inverse relationships with SV2A measures across diagnoses, and initial findings also suggest transdiagnostic relationships with mood and anxiety symptoms. This suggests that synaptic abnormalities could be a common pathophysiological substrate underlying cognitive and, potentially, affective symptoms. We consider limitations of evidence and future directions; highlighting the need to develop postsynaptic imaging markers and for longitudinal studies to test causal mechanisms.
Collapse
Affiliation(s)
- Oliver Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England.
- South London & the Maudsley NHS Trust, London, England.
- London Institute of Medical Sciences, London, England.
| | - Julia Marcinkowska
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Richard Carr
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
- South London & the Maudsley NHS Trust, London, England
- London Institute of Medical Sciences, London, England
| |
Collapse
|
5
|
Xiong M, Lubberink M, Appel L, Fang XT, Danfors T, Kumlien E, Antoni G. Evaluation of [ 11C]UCB-A positron emission tomography in human brains. EJNMMI Res 2024; 14:56. [PMID: 38884834 PMCID: PMC11183037 DOI: 10.1186/s13550-024-01117-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND In preclinical studies, the positron emission tomography (PET) imaging with [11C]UCB-A provided promising results for imaging synaptic vesicle protein 2A (SV2A) as a proxy for synaptic density. This paper reports the first-in-human [11C]UCB-A PET study to characterise its kinetics in healthy subjects and further evaluate SV2A-specific binding. RESULTS Twelve healthy subjects underwent 90-min baseline [11C]UCB-A scans with PET/MRI, with two subjects participating in an additional blocking scan with the same scanning procedure after a single dose of levetiracetam (1500 mg). Our results indicated abundant [11C]UCB-A brain uptake across all cortical regions, with slow elimination. Kinetic modelling of [11C]UCB-A PET using various compartment models suggested that the irreversible two-tissue compartment model best describes the kinetics of the radioactive tracer. Accordingly, the Patlak graphical analysis was used to simplify the analysis. The estimated SV2A occupancy determined by the Lassen plot was around 66%. Significant specific binding at baseline and comparable binding reduction as grey matter precludes the use of centrum semiovale as reference tissue. CONCLUSIONS [11C]UCB-A PET imaging enables quantifying SV2A in vivo. However, its slow kinetics require a long scan duration, which is impractical with the short half-life of carbon-11. Consequently, the slow kinetics and complicated quantification methods may restrict its use in humans.
Collapse
Affiliation(s)
- Mengfei Xiong
- Molecular Imaging and Medical Physics, Department of Surgical Sciences, Uppsala University, Entrance 70, 75185, Uppsala, Sweden.
| | - Mark Lubberink
- Molecular Imaging and Medical Physics, Department of Surgical Sciences, Uppsala University, Entrance 70, 75185, Uppsala, Sweden
| | - Lieuwe Appel
- Molecular Imaging and Medical Physics, Department of Surgical Sciences, Uppsala University, Entrance 70, 75185, Uppsala, Sweden
| | - Xiaotian Tsong Fang
- Molecular Imaging and Medical Physics, Department of Surgical Sciences, Uppsala University, Entrance 70, 75185, Uppsala, Sweden
- Julius Clinical BV, Zeist, The Netherlands
| | - Torsten Danfors
- Molecular Imaging and Medical Physics, Department of Surgical Sciences, Uppsala University, Entrance 70, 75185, Uppsala, Sweden
| | - Eva Kumlien
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gunnar Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Bavarsad MS, Grinberg LT. SV2A PET imaging in human neurodegenerative diseases. Front Aging Neurosci 2024; 16:1380561. [PMID: 38699560 PMCID: PMC11064927 DOI: 10.3389/fnagi.2024.1380561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
This manuscript presents a thorough review of synaptic vesicle glycoprotein 2A (SV2A) as a biomarker for synaptic integrity using Positron Emission Tomography (PET) in neurodegenerative diseases. Synaptic pathology, characterized by synaptic loss, has been linked to various brain diseases. Therefore, there is a need for a minimally invasive approach to measuring synaptic density in living human patients. Several radiotracers targeting synaptic vesicle protein 2A (SV2A) have been created and effectively adapted for use in human subjects through PET scans. SV2A is an integral glycoprotein found in the membranes of synaptic vesicles in all synaptic terminals and is widely distributed throughout the brain. The review delves into the development of SV2A-specific PET radiotracers, highlighting their advancements and limitations in neurodegenerative diseases. Among these tracers, 11C-UCB-J is the most used so far. We summarize and discuss an increasing body of research that compares measurements of synaptic density using SV2A PET with other established indicators of neurodegenerative diseases, including cognitive performance and radiological findings, thus providing a comprehensive analysis of SV2A's effectiveness and reliability as a diagnostic tool in contrast to traditional markers. Although the literature overall suggests the promise of SV2A as a diagnostic and therapeutic monitoring tool, uncertainties persist regarding the superiority of SV2A as a biomarker compared to other available markers. The review also underscores the paucity of studies characterizing SV2A distribution and loss in human brain tissue from patients with neurodegenerative diseases, emphasizing the need to generate quantitative neuropathological maps of SV2A density in cases with neurodegenerative diseases to fully harness the potential of SV2A PET imaging in clinical settings. We conclude by outlining future research directions, stressing the importance of integrating SV2A PET imaging with other biomarkers and clinical assessments and the need for longitudinal studies to track SV2A changes throughout neurodegenerative disease progression, which could lead to breakthroughs in early diagnosis and the evaluation of new treatments.
Collapse
Affiliation(s)
| | - Lea T. Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
7
|
Rossi R, Bærentzen SL, Thomsen MB, Real CC, Wegener G, Grassi-Oliveira R, Gjedde A, Landau AM. A single dose of cocaine raises SV2A density in hippocampus of adolescent rats. Acta Neuropsychiatr 2024; 36:109-117. [PMID: 36847240 DOI: 10.1017/neu.2023.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
OBJECTIVE Cocaine is a highly addictive psychostimulant that affects synaptic activity with structural and functional adaptations of neurons. The transmembrane synaptic vesicle glycoprotein 2A (SV2A) of pre-synaptic vesicles is commonly used to measure synaptic density, as a novel approach to the detection of synaptic changes. We do not know if a single dose of cocaine suffices to affect pre-synaptic SV2A density, especially during adolescence when synapses undergo intense maturation. Here, we explored potential changes of pre-synaptic SV2A density in target brain areas associated with the cocaine-induced boost of dopaminergic neurotransmission, specifically testing if the effects would last after the return of dopamine levels to baseline. METHODS We administered cocaine (20 mg/kg i.p.) or saline to rats in early adolescence, tested their activity levels and removed the brains 1 hour and 7 days after injection. To evaluate immediate and lasting effects, we did autoradiography with [3H]UCB-J, a specific tracer for SV2A, in medial prefrontal cortex, striatum, nucleus accumbens, amygdala, and dorsal and ventral areas of hippocampus. We also measured the striatal binding of [3H]GBR-12935 to test cocaine's occupancy of the dopamine transporter at both times of study. RESULTS We found a significant increase of [3H]UCB-J binding in the dorsal and ventral sections of hippocampus 7 days after the cocaine administration compared to saline-injected rats, but no differences 1 hour after the injection. The [3H]GBR-12935 binding remained unchanged at both times. CONCLUSION Cocaine provoked lasting changes of hippocampal synaptic SV2A density after a single exposure during adolescence.
Collapse
Affiliation(s)
- Rachele Rossi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Simone Larsen Bærentzen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Majken B Thomsen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Caroline C Real
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Rodrigo Grassi-Oliveira
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
- Department of Neuroscience, University of Copenhagen, Denmark
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
8
|
Visser M, O'Brien JT, Mak E. In vivo imaging of synaptic density in neurodegenerative disorders with positron emission tomography: A systematic review. Ageing Res Rev 2024; 94:102197. [PMID: 38266660 DOI: 10.1016/j.arr.2024.102197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Positron emission tomography (PET) with radiotracers that bind to synaptic vesicle glycoprotein 2 A (SV2A) enables quantification of synaptic density in the living human brain. Assessing the regional distribution and severity of synaptic density loss will contribute to our understanding of the pathological processes that precede atrophy in neurodegeneration. In this systematic review, we provide a discussion of in vivo SV2A PET imaging research for quantitative assessment of synaptic density in various dementia conditions: amnestic Mild Cognitive Impairment and Alzheimer's disease, Frontotemporal dementia, Progressive supranuclear palsy and Corticobasal degeneration, Parkinson's disease and Dementia with Lewy bodies, Huntington's disease, and Spinocerebellar Ataxia. We discuss the main findings concerning group differences and clinical-cognitive correlations, and explore relations between SV2A PET and other markers of pathology. Additionally, we touch upon synaptic density in healthy ageing and outcomes of radiotracer validation studies. Studies were identified on PubMed and Embase between 2018 and 2023; last searched on the 3rd of July 2023. A total of 36 studies were included, comprising 5 on normal ageing, 21 clinical studies, and 10 validation studies. Extracted study characteristics were participant details, methodological aspects, and critical findings. In summary, the small but growing literature on in vivo SV2A PET has revealed different spatial patterns of synaptic density loss among various neurodegenerative disorders that correlate with cognitive functioning, supporting the potential role of SV2A PET imaging for differential diagnosis. SV2A PET imaging shows tremendous capability to provide novel insights into the aetiology of neurodegenerative disorders and great promise as a biomarker for synaptic density reduction. Novel directions for future synaptic density research are proposed, including (a) longitudinal imaging in larger patient cohorts of preclinical dementias, (b) multi-modal mapping of synaptic density loss onto other pathological processes, and (c) monitoring therapeutic responses and assessing drug efficacy in clinical trials.
Collapse
Affiliation(s)
- Malouke Visser
- Department of Psychiatry, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, United Kingdom; Neuropsychology and Rehabilitation Psychology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - John T O'Brien
- Department of Psychiatry, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, United Kingdom
| | - Elijah Mak
- Department of Psychiatry, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, United Kingdom.
| |
Collapse
|
9
|
Desmond KL, Lindberg A, Garcia A, Tong J, Harkness MB, Dobrota E, Smart K, Uribe C, Meyer JH, Houle S, Strafella AP, Li S, Huang Y, Vasdev N. First-in-Human PET Imaging of [ 18F]SDM-4MP3: A Cautionary Tale. Mol Imaging 2023; 2023:8826977. [PMID: 37719326 PMCID: PMC10504053 DOI: 10.1155/2023/8826977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/06/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
[18F]SynVesT-1 is a PET radiopharmaceutical that binds to the synaptic vesicle protein 2A (SV2A) and serves as a biomarker of synaptic density with widespread clinical research applications in psychiatry and neurodegeneration. The initial goal of this study was to concurrently conduct PET imaging studies with [18F]SynVesT-1 at our laboratories. However, the data in the first two human PET studies had anomalous biodistribution despite the injected product meeting all specifications during the prerelease quality control protocols. Further investigation, including imaging in rats as well as proton and carbon 2D-NMR spectroscopic studies, led to the discovery that a derivative of the precursor had been received from the manufacturer. Hence, we report our investigation and the first-in-human study of [18F]SDM-4MP3, a structural variant of [18F]SynVesT-1, which does not have the requisite characteristics as a PET radiopharmaceutical for imaging SV2A in the central nervous system.
Collapse
Affiliation(s)
- Kimberly L. Desmond
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Ontario, Canada
| | - Anton Lindberg
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Armando Garcia
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Michael B. Harkness
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Elena Dobrota
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Kelly Smart
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Ontario, Canada
| | - Carme Uribe
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Jeffrey H. Meyer
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Ontario, Canada
| | - Sylvain Houle
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Ontario, Canada
| | - Antonio P. Strafella
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Krembil Brain Institute, University Health Network, University of Toronto, Ontario, Canada
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry & Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Ontario, Canada
| |
Collapse
|
10
|
Singh P, Singh D, Srivastava P, Mishra G, Tiwari AK. Evaluation of advanced, pathophysiologic new targets for imaging of CNS. Drug Dev Res 2023; 84:484-513. [PMID: 36779375 DOI: 10.1002/ddr.22040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/12/2022] [Accepted: 12/31/2022] [Indexed: 02/14/2023]
Abstract
The inadequate information about the in vivo pathological, physiological, and neurological impairments, as well as the absence of in vivo tools for assessing brain penetrance and the efficiency of newly designed drugs, has hampered the development of new techniques for the treatment for variety of new central nervous system (CNS) diseases. The searching sites such as Science Direct and PubMed were used to find out the numerous distinct tracers across 16 CNS targets including tau, synaptic vesicle glycoprotein, the adenosine 2A receptor, the phosphodiesterase enzyme PDE10A, and the purinoceptor, among others. Among the most encouraging are [18 F]FIMX for mGluR imaging, [11 C]Martinostat for Histone deacetylase, [18 F]MNI-444 for adenosine 2A imaging, [11 C]ER176 for translocator protein, and [18 F]MK-6240 for tau imaging. We also reviewed the findings for each tracer's features and potential for application in CNS pathophysiology and therapeutic evaluation investigations, including target specificity, binding efficacy, and pharmacokinetic factors. This review aims to present a current evaluation of modern positron emission tomography tracers for CNS targets, with a focus on recent advances for targets that have newly emerged for imaging in humans.
Collapse
Affiliation(s)
- Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Deepika Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Pooja Srivastava
- Division of Cyclotron and Radiopharmaceuticals Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Gauri Mishra
- Department of Zoology, Swami Shraddhananad College, University of Delhi, Alipur, Delhi, India
| | - Anjani K Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
11
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
McErlain H, McLean EB, Morgan TEF, Burianova VK, Tavares AAS, Sutherland A. Organocatalytic Asymmetric Synthesis of SynVesT-1, a Synaptic Density Positron Emission Tomography Imaging Agent. J Org Chem 2022; 87:14443-14451. [PMID: 36222243 PMCID: PMC9639009 DOI: 10.1021/acs.joc.2c01895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Heterocyclic nonacetamide ligands are used as positron emission tomography (PET) imaging agents of the synaptic vesicle glycoprotein 2A (SV2A), with potential applications in the diagnosis of various neuropsychiatric diseases. To date, the main synthetic strategy to access these optically active compounds has involved the racemic synthesis of a late-stage intermediate followed by the separation of the enantiomers. Here, we describe the use of iminium organocatalysis for the asymmetric synthesis of SynVesT-1, an important PET imaging agent of SV2A. The key step involved the conjugate addition of nitromethane with a cinnamaldehyde in the presence of the Jørgensen-Hayashi catalyst using the Merck dual acid cocatalyst system. Pinnick-type oxidation and esterification of the adduct was then followed by chemoselective nitro group reduction and cyclization using nickel borate. N-Alkylation of the resulting lactam then completed the seven-step synthesis of SynVesT-1. This approach was amenable for the synthesis of an organotin analogue, which following copper(II)-mediated fluoro-destannylation allowed rapid access to [18F]SynVesT-1.
Collapse
Affiliation(s)
- Holly McErlain
- School
of Chemistry, The Joseph Black Building, University of Glasgow, GlasgowG12 8QQ, U.K.
| | - Euan B. McLean
- School
of Chemistry, The Joseph Black Building, University of Glasgow, GlasgowG12 8QQ, U.K.
| | - Timaeus E. F. Morgan
- BHF-University
Centre for Cardiovascular Science, University
of Edinburgh, EdinburghEH16 4TJ, U.K.
| | - Valeria K. Burianova
- School
of Chemistry, The Joseph Black Building, University of Glasgow, GlasgowG12 8QQ, U.K.
| | - Adriana A. S. Tavares
- BHF-University
Centre for Cardiovascular Science, University
of Edinburgh, EdinburghEH16 4TJ, U.K.
| | - Andrew Sutherland
- School
of Chemistry, The Joseph Black Building, University of Glasgow, GlasgowG12 8QQ, U.K.,
| |
Collapse
|
13
|
Carson RE, Naganawa M, Toyonaga T, Koohsari S, Yang Y, Chen MK, Matuskey D, Finnema SJ. Imaging of Synaptic Density in Neurodegenerative Disorders. J Nucl Med 2022; 63:60S-67S. [PMID: 35649655 DOI: 10.2967/jnumed.121.263201] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/10/2022] [Indexed: 02/07/2023] Open
Abstract
PET technology has produced many radiopharmaceuticals that target specific brain proteins and other measures of brain function. Recently, a new approach has emerged to image synaptic density by targeting the synaptic vesicle protein 2A (SV2A), an integral glycoprotein in the membrane of synaptic vesicles and widely distributed throughout the brain. Multiple SV2A ligands have been developed and translated to human use. The most successful of these to date is 11C-UCB-J, because of its high uptake, moderate metabolism, and effective quantification with a 1-tissue-compartment model. Further, since SV2A is the target of the antiepileptic drug levetiracetam, human blocking studies have characterized specific binding and potential reference regions. Regional brain SV2A levels were shown to correlate with those of synaptophysin, another commonly used marker of synaptic density, providing the basis for SV2A PET imaging to have broad utility across neuropathologic diseases. In this review, we highlight the development of SV2A tracers and the evaluation of quantification methods, including compartment modeling and simple tissue ratios. Mouse and rat models of neurodegenerative diseases have been studied with small-animal PET, providing validation by comparison to direct tissue measures. Next, we review human PET imaging results in multiple neurodegenerative disorders. Studies on Parkinson disease and Alzheimer disease have progressed most rapidly at multiple centers, with generally consistent results of patterns of SV2A or synaptic loss. In Alzheimer disease, the synaptic loss patterns differ from those of amyloid, tau, and 18F-FDG, although intertracer and interregional correlations have been found. Smaller studies have been reported in other disorders, including Lewy body dementia, frontotemporal dementia, Huntington disease, progressive supranuclear palsy, and corticobasal degeneration. In conclusion, PET imaging of SV2A has rapidly developed, and qualified radioligands are available. PET studies on humans indicate that SV2A loss might be specific to disease-associated brain regions and consistent with synaptic density loss. The recent availability of new 18F tracers, 18F-SynVesT-1 and 18F-SynVesT-2, will substantially broaden the application of SV2A PET. Future studies are needed in larger patient cohorts to establish the clinical value of SV2A PET and its potential for diagnosis and progression monitoring of neurodegenerative diseases, as well as efficacy assessment of disease-modifying therapies.
Collapse
Affiliation(s)
- Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Sheida Koohsari
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Yanghong Yang
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut; and
| | - Sjoerd J Finnema
- Neuroscience Discovery Research, Translational Imaging, AbbVie, North Chicago, Illinois
| |
Collapse
|
14
|
Rossi R, Arjmand S, Bærentzen SL, Gjedde A, Landau AM. Synaptic Vesicle Glycoprotein 2A: Features and Functions. Front Neurosci 2022; 16:864514. [PMID: 35573314 PMCID: PMC9096842 DOI: 10.3389/fnins.2022.864514] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/05/2022] [Indexed: 01/05/2023] Open
Abstract
In recent years, the field of neuroimaging dramatically moved forward by means of the expeditious development of specific radioligands of novel targets. Among these targets, the synaptic vesicle glycoprotein 2A (SV2A) is a transmembrane protein of synaptic vesicles, present in all synaptic terminals, irrespective of neurotransmitter content. It is involved in key functions of neurons, focused on the regulation of neurotransmitter release. The ubiquitous expression in gray matter regions of the brain is the basis of its candidacy as a marker of synaptic density. Following the development of molecules derived from the structure of the anti-epileptic drug levetiracetam, which selectively binds to SV2A, several radiolabeled markers have been synthetized to allow the study of SV2A distribution with positron emission tomography (PET). These radioligands permit the evaluation of in vivo changes of SV2A distribution held to be a potential measure of synaptic density in physiological and pathological conditions. The use of SV2A as a biomarker of synaptic density raises important questions. Despite numerous studies over the last decades, the biological function and the expressional properties of SV2A remain poorly understood. Some functions of SV2A were claimed, but have not been fully elucidated. While the expression of SV2A is ubiquitous, stronger associations between SV2A and Υ amino butyric acid (GABA)-ergic rather than glutamatergic synapses were observed in some brain structures. A further issue is the unclear interaction between SV2A and its tracers, which reflects a need to clarify what really is detected with neuroimaging tools. Here, we summarize the current knowledge of the SV2A protein and we discuss uncertain aspects of SV2A biology and physiology. As SV2A expression is ubiquitous, but likely more strongly related to a certain type of neurotransmission in particular circumstances, a more extensive knowledge of the protein would greatly facilitate the analysis and interpretation of neuroimaging results by allowing the evaluation not only of an increase or decrease of the protein level, but also of the type of neurotransmission involved.
Collapse
Affiliation(s)
- Rachele Rossi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Simone Larsen Bærentzen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
16
|
Jiang Z, Cheng X, Chen H, Zheng W, Sun Y, Yu Z, Yang T, Zhang L, Fan D, Yang Z, Liu Y, Ai L, Wu Z. [ 18F]BIBD-181: A Novel Positron Emission Tomography Tracer Specific for Synaptic Vesicle Glycoprotein 2A. ACS Med Chem Lett 2022; 13:720-726. [PMID: 35450380 PMCID: PMC9014511 DOI: 10.1021/acsmedchemlett.2c00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/28/2022] [Indexed: 11/28/2022] Open
Abstract
Dysfunction or decreased expression of synaptic vesicle glycoprotein 2A (SV2A) is closely related to the progression of neurodegenerative diseases and psychiatric disorders. The development of positron emission tomography (PET) tracers targeting SV2A can provide a strong imaging basis for the diagnosis and treatment of these diseases. Herein we report the synthesis of the novel radiotracer [18F]BIBD-181 and its preclinical evaluation. The absolute configuration of BIBD-181 was confirmed by the single-crystal structure of its precursor. The in vitro binding assay of BIBD-181 showed high SV2A binding affinity. Compared with previously reported tracers, [18F]BIBD-181 has mild labeling conditions, simple operation, and high yield. The in vivo metabolism of [18F]BIBD-181 is similar to that of UCB derivatives, and the metabolites do not interfere with brain PET imaging. Biodistribution and PET studies showed that [18F]BIBD-181 has high brain uptake and good pharmacokinetics. Autoradiography and PET inhibition studies indicated that [18F]BIBD-181 specifically binds SV2A. Because [18F]BIBD-181 exhibits excellent properties, it may be a reliable probe of quantities for SV2A-related disease diagnosis.
Collapse
Affiliation(s)
- Zeng Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xuebo Cheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Wei Zheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yuli Sun
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ziyue Yu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Tingyu Yang
- School of Pharmaceutical Science, Capital Medical University, Beijing 100069, China
| | - Lu Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Di Fan
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100069, China
| | - Zhihao Yang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100069, China
| | - Yajing Liu
- School of Pharmaceutical Science, Capital Medical University, Beijing 100069, China
| | - Lin Ai
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100069, China
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
17
|
Li W, Lv L, Luo XJ. In vivo study sheds new light on the dendritic spine pathology hypothesis of schizophrenia. Mol Psychiatry 2022; 27:1866-1868. [PMID: 35079121 DOI: 10.1038/s41380-022-01449-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan, 453002, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China. .,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China.
| |
Collapse
|
18
|
Assessment of the In Vivo Relationship Between Cerebral Hypometabolism, Tau Deposition, TSPO Expression, and Synaptic Density in a Tauopathy Mouse Model: a Multi-tracer PET Study. Mol Neurobiol 2022; 59:3402-3413. [PMID: 35312967 PMCID: PMC9148291 DOI: 10.1007/s12035-022-02793-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/05/2022] [Indexed: 11/03/2022]
Abstract
Cerebral glucose hypometabolism is a typical hallmark of Alzheimer’s disease (AD), usually associated with ongoing neurodegeneration and neuronal dysfunction. However, underlying pathological processes are not fully understood and reproducibility in animal models is not well established. The aim of the present study was to investigate the regional interrelation of glucose hypometabolism measured by [18F]FDG positron emission tomography (PET) with various molecular targets of AD pathophysiology using the PET tracers [18F]PI-2620 for tau deposition, [18F]DPA-714 for TSPO expression associated with neuroinflammation, and [18F]UCB-H for synaptic density in a transgenic tauopathy mouse model. Seven-month-old rTg4510 mice (n = 8) and non-transgenic littermates (n = 8) were examined in a small animal PET scanner with the tracers listed above. Hypometabolism was observed throughout the forebrain of rTg4510 mice. Tau pathology, increased TSPO expression, and synaptic loss were co-localized in the cortex and hippocampus and correlated with hypometabolism. In the thalamus, however, hypometabolism occurred in the absence of tau-related pathology. Thus, cerebral hypometabolism was associated with two regionally distinct forms of molecular pathology: (1) characteristic neuropathology of the Alzheimer-type including synaptic degeneration and neuroinflammation co-localized with tau deposition in the cerebral cortex, and (2) pathological changes in the thalamus in the absence of other markers of AD pathophysiology, possibly reflecting downstream or remote adaptive processes which may affect functional connectivity. Our study demonstrates the feasibility of a multitracer approach to explore complex interactions of distinct AD-pathomechanisms in vivo in a small animal model. The observations demonstrate that multiple, spatially heterogeneous pathomechanisms can contribute to hypometabolism observed in AD mouse models and they motivate future longitudinal studies as well as the investigation of possibly comparable pathomechanisms in human patients.
Collapse
|
19
|
Angarita GA, Worhunsky PD, Naganawa M, Toyonaga T, Nabulsi NB, Li CSR, Esterlis I, Skosnik PD, Radhakrishnan R, Pittman B, Gueorguieva R, Potenza MN, Finnema SJ, Huang Y, Carson RE, Malison RT. Lower prefrontal cortical synaptic vesicle binding in cocaine use disorder: An exploratory 11 C-UCB-J positron emission tomography study in humans. Addict Biol 2022; 27:e13123. [PMID: 34852401 PMCID: PMC8891080 DOI: 10.1111/adb.13123] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/22/2021] [Accepted: 11/12/2021] [Indexed: 12/22/2022]
Abstract
Preclinical studies have revealed robust and long-lasting alterations in dendritic spines in the brain following cocaine exposure. Such alterations are hypothesized to underlie enduring maladaptive behaviours observed in cocaine use disorder (CUD). The current study explored whether synaptic density is altered in CUD. Fifteen individuals with DSM-5 CUD and 15 demographically matched healthy control (HC) subjects participated in a single 11 C-UCB-J positron emission tomography scan to assess density of synaptic vesicle glycoprotein 2A (SV2A). The volume of distribution (VT ) and the plasma-free fraction-corrected form of the total volume of distribution (VT /fP ) were analysed in the anterior cingulate cortex (ACC), dorsomedial and ventromedial prefrontal cortex (PFC), lateral and medial orbitofrontal cortex (OFC) and ventral striatum. A significant diagnostic-group-by-region interaction was observed for VT and VT /fP . Post hoc analyses revealed no differences on VT , while for VT /fP showed lower values in CUD as compared with HC subjects in the ACC (-10.9%, p = 0.02), ventromedial PFC (-9.9%, p = 0.02) and medial OFC (-9.9%, p = 0.04). Regional VT /fP values in CUD, though unrelated to measures of lifetime cocaine use, were positively correlated with the frequency of recent cocaine use (p = 0.02-0.03) and negatively correlated with cocaine abstinence (p = 0.008-0.03). These findings provide initial preliminary in vivo evidence of altered (lower) synaptic density in the PFC of humans with CUD. Cross-sectional variation in SV2A availability as a function of recent cocaine use and abstinence suggests that synaptic density may be dynamically and plastically regulated by acute cocaine, an observation that merits direct testing by studies using more definitive longitudinal designs.
Collapse
Affiliation(s)
- Gustavo A. Angarita
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
| | - Patrick D. Worhunsky
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Nabeel B. Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Chiang-Shan R. Li
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
| | - Irina Esterlis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Patrick D. Skosnik
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Rajiv Radhakrishnan
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Brian Pittman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
| | - Ralitza Gueorguieva
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven CT 06520
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
- Connecticut Council on Problem Gambling, 100 Great Meadow Road, Wethersfield, CT 06109
- Child Study Center, Yale University School of Medicine, 234 South Frontage Road, New Haven, CT 06510
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510
| | - Sjoerd J. Finnema
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Ave, New Haven, CT 06519
| | - Robert T. Malison
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite # 901, New Haven, CT 06511
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519
| |
Collapse
|
20
|
Wang T, Lin Q, Zhang Y, Xu Z, Shi D, Cheng Y, Fu Z, Tan H, Cheng D, Shi H. Synthesis and biological evaluation of novel PET tracers [ 18F]AG120 & [ 18F]AG135 for imaging mutant isocitrate dehydrogenase 1 expression. Bioorg Med Chem 2022; 53:116525. [PMID: 34871844 DOI: 10.1016/j.bmc.2021.116525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/04/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
Abstract
Mutations in isocitrate dehydrogenase 1 (IDH1) are commonly found in various human malignancies. Inhibitors of several mutant IDH1 enzymes have entered clinical trials as target therapeutic drugs for the treatment of patients with IDH1 mutations. Herein, we report the synthesis and evaluation of two 18F-labeled tracers, [18F]AG120 and [18F]AG135 for imaging expression of mutated IDH1 in positron emission tomography (PET). [18F]AG120 and [18F]AG135 were synthesized in decay-corrected radiochemical yield of 1 % and 3 %, respectively, high molar activity (52-66 MBq/nmol and 216-339 MBq/nmol, respectively) and high radiochemical purity (>99%). Both tracers showed good in vitro stability, selective uptake into mutated IDH1-expressing cells and good pharmacokinetic profiles with low uptake in most organs/tissues. Furthermore, [18F]AG120 micro-PET/CT imaging displayed significantly greater uptake in IDH1-mutant than in wild-type tumors, Relatively, uptake of [18F]AG135 was observed neither in IDH1-mutant tumor xenografts nor in wild-type tumors. This study suggests that [18F]AG120 is a promising radiotracer for PET imaging of IDH1 mutation, However, further optimization and investigation are necessary for [18F]AG135 due to the limited uptake in mutated IDH1-expressing tumors.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Qingyu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China; Institute of Nuclear Medicine, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China
| | - Yingying Zhang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Zhan Xu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Dai Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Yuan Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Zhequan Fu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Hui Tan
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China; Institute of Nuclear Medicine, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai 200032, China; Institute of Nuclear Medicine, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China.
| |
Collapse
|
21
|
Brumberg J, Varrone A. New PET radiopharmaceuticals for imaging CNS diseases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
22
|
Kong Y, Zhang S, Huang L, Zhang C, Xie F, Zhang Z, Huang Q, Jiang D, Li J, Zhou W, Hua T, Sun B, Wang J, Guan Y. Positron Emission Computed Tomography Imaging of Synaptic Vesicle Glycoprotein 2A in Alzheimer's Disease. Front Aging Neurosci 2021; 13:731114. [PMID: 34795573 PMCID: PMC8593388 DOI: 10.3389/fnagi.2021.731114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder seen in age-dependent dementia. There is currently no effective treatment for AD, which may be attributed in part to lack of a clear underlying mechanism. Early diagnosis of AD is of great significance to control the development of the disease. Synaptic loss is an important pathology in the early stage of AD, therefore the measurement of synaptic density using molecular imaging technology may be an effective way to early diagnosis of AD. Synaptic vesicle glycoprotein 2A (SV2A) is located in the presynaptic vesicle membrane of virtually all synapses. SV2A Positron Emission Computed Tomography (PET) could provide a way to measure synaptic density quantitatively in living humans and to track changes in synaptic density in AD. In view of the fact that synaptic loss is the pathology of both epilepsy and AD, this review summarizes the potential role of SV2A in the pathogenesis of AD, and suggests that SV2A should be used as an important target molecule of PET imaging agent for the early diagnosis of AD.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Shibo Zhang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Lin Huang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhengwei Zhang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Donglang Jiang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Junpeng Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiyan Zhou
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Tao Hua
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Pracitto R, Wilcox KC, Lindemann M, Tong J, Zheng C, Li S, Finnema SJ, Huang Y, Cai Z. Further Investigation of Synaptic Vesicle Protein 2A (SV2A) Ligands Designed for Positron Emission Tomography and Single-Photon Emission Computed Tomography Imaging: Synthesis and Structure-Activity Relationship of Substituted Pyridinylmethyl-4-(3,5-difluorophenyl)pyrrolidin-2-ones. ACS OMEGA 2021; 6:27676-27683. [PMID: 34722967 PMCID: PMC8552234 DOI: 10.1021/acsomega.1c02433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/22/2021] [Indexed: 06/13/2023]
Abstract
A series of synaptic vesicle protein 2A (SV2A) ligands were synthesized to explore the structure-activity relationship and to help further investigate a hydrogen bonding pharmacophore hypothesis. Racemic SynVesT-1 was used as a lead compound to explore the replacement of the 3-methyl group on the pyridinyl moiety with halogens and hydrocarbons. Pyridinyl isomers of racemic SynVesT-1 were also investigated. Highly potent analogs were discovered including a 3-iodo pyridinyl ligand amenable to investigation as a PET or SPECT imaging agent.
Collapse
Affiliation(s)
- Richard Pracitto
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Kyle C. Wilcox
- Translational
Imaging Neuroscience, AbbVie, North Chicago, Illinois 60064, United States
| | - Marcel Lindemann
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Jie Tong
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Chao Zheng
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Songye Li
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Sjoerd J. Finnema
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
- Translational
Imaging Neuroscience, AbbVie, North Chicago, Illinois 60064, United States
| | - Yiyun Huang
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Zhengxin Cai
- PET
Center, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| |
Collapse
|
24
|
Tuncel H, Boellaard R, Coomans EM, de Vries EFJ, Glaudemans AWJM, Feltes PK, García DV, Verfaillie SCJ, Wolters EE, Sweeney SP, Ryan JM, Ivarsson M, Lynch BA, Schober P, Scheltens P, Schuit RC, Windhorst AD, De Deyn PP, van Berckel BNM, Golla SSV. Kinetics and 28-day test-retest repeatability and reproducibility of [ 11C]UCB-J PET brain imaging. J Cereb Blood Flow Metab 2021; 41:1338-1350. [PMID: 34013797 PMCID: PMC8138337 DOI: 10.1177/0271678x20964248] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/19/2020] [Accepted: 09/27/2020] [Indexed: 11/18/2022]
Abstract
[11C]UCB-J is a novel radioligand that binds to synaptic vesicle glycoprotein 2A (SV2A). The main objective of this study was to determine the 28-day test-retest repeatability (TRT) of quantitative [11C]UCB-J brain positron emission tomography (PET) imaging in Alzheimer's disease (AD) patients and healthy controls (HCs). Nine HCs and eight AD patients underwent two 60 min dynamic [11C]UCB-J PET scans with arterial sampling with an interval of 28 days. The optimal tracer kinetic model was assessed using the Akaike criteria (AIC). Micro-/macro-parameters such as tracer delivery (K1) and volume of distribution (VT) were estimated using the optimal model. Data were also analysed for simplified reference tissue model (SRTM) with centrum semi-ovale (white matter) as reference region. Based on AIC, both 1T2k_VB and 2T4k_VB described the [11C]UCB-J kinetics equally well. Analysis showed that whole-brain grey matter TRT for VT, DVR and SRTM BPND were -2.2% ± 8.5, 0.4% ± 12.0 and -8.0% ± 10.2, averaged over all subjects. [11C]UCB-J kinetics can be well described by a 1T2k_VB model, and a 60 min scan duration was sufficient to obtain reliable estimates for both plasma input and reference tissue models. TRT for VT, DVR and BPND was <15% (1SD) averaged over all subjects and indicates adequate quantitative repeatability of [11C]UCB-J PET.
Collapse
Affiliation(s)
- Hayel Tuncel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Emma M Coomans
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Erik FJ de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - Andor WJM Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - Paula Kopschina Feltes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - David V García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center, University of Groningen, Groningen, The Netherlands
| | - Sander CJ Verfaillie
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Emma E Wolters
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | | | | | | | | | - Patrick Schober
- Department of Anaesthesiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Peter P De Deyn
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Alzheimer Research Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart NM van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sandeep SV Golla
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Sadasivam P, Fang XT, Toyonaga T, Lee S, Xu Y, Zheng MQ, Spurrier J, Huang Y, Strittmatter SM, Carson RE, Cai Z. Quantification of SV2A Binding in Rodent Brain Using [ 18F]SynVesT-1 and PET Imaging. Mol Imaging Biol 2021; 23:372-381. [PMID: 33258040 PMCID: PMC8105262 DOI: 10.1007/s11307-020-01567-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Synapse loss is a hallmark of Alzheimer's disease (AD) and correlates with cognitive decline. The validation of a noninvasive in vivo imaging approach to quantify synapse would greatly facilitate our understanding of AD pathogenesis and assist drug developments for AD. As animal models of neurodegenerative and neuropsychiatric disorders play a critical role in the drug discovery and development process, a robust, objective, and translational method for quantifying therapeutic drug efficacy in animal models will facilitate the drug development process. In this study, we tested the quantification reliability of the SV2A PET tracer, [18F]SynVesT-1, in a mouse model of AD (APP/PS1) and wild-type controls, and developed a simplified quantification method to facilitate large cohort preclinical imaging studies. PROCEDURES We generated nondisplaceable binding potential (BPND) and distribution volume ratio (DVR) values using the simplified reference tissue model (SRTM) on the 90-min dynamic PET imaging data, with brain stem and cerebellum as the reference region, respectively. Then, we correlated the standardized uptake value ratio (SUVR)-1 and SUVR averaged from different imaging windows with BPND and DVR, using brain stem and cerebellum as the reference region, respectively. We performed homologous competitive binding assay and autoradiographic saturation binding assay using [18F]SynVesT-1 to calculate the Bmax and Kd. RESULTS Using brain stem as the reference region, the averaged SUVR-1 from 30 to 60 min postinjection correlated well with the BPND calculated using SRTM. Using cerebellum as the reference region, the averaged SUVR from 30 to 60 min postinjection correlated well with the SRTM DVR. From the homologous competitive binding assay and autoradiographic saturation binding assay, the calculated the Bmax and Kd were 4.5-18 pmol/mg protein and 9.8-19.6 nM, respectively, for rodent brain tissue. CONCLUSIONS This simplified SUVR method provides reasonable SV2A measures in APP/PS1 mice and their littermate controls. Our data indicate that, in lieu of a full 90-min dynamic scan, a 30-min static PET scan (from 30 to 60 min postinjection) would be sufficient to provide quantification data on SV2A expression, equivalent to the data generated from kinetic modeling. The methods developed here are readily applicable to the evaluation of therapeutic effects of novel drugs in this rodent model using [18F]SynVesT-1 and small animal PET.
Collapse
Affiliation(s)
- Pragalath Sadasivam
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Xiaotian T. Fang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Takuya Toyonaga
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Supum Lee
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Yuping Xu
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Ming-Qiang Zheng
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Joshua Spurrier
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Cell Biology, Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Yiyun Huang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Stephen M. Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Cell Biology, Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Richard E. Carson
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Zhengxin Cai
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Bao W, Xie F, Zuo C, Guan Y, Huang YH. PET Neuroimaging of Alzheimer's Disease: Radiotracers and Their Utility in Clinical Research. Front Aging Neurosci 2021; 13:624330. [PMID: 34025386 PMCID: PMC8134674 DOI: 10.3389/fnagi.2021.624330] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's Disease (AD), the leading cause of senile dementia, is a progressive neurodegenerative disorder affecting millions of people worldwide and exerting tremendous socioeconomic burden on all societies. Although definitive diagnosis of AD is often made in the presence of clinical manifestations in late stages, it is now universally believed that AD is a continuum of disease commencing from the preclinical stage with typical neuropathological alterations appearing decades prior to its first symptom, to the prodromal stage with slight symptoms of amnesia (amnestic mild cognitive impairment, aMCI), and then to the terminal stage with extensive loss of basic cognitive functions, i.e., AD-dementia. Positron emission tomography (PET) radiotracers have been developed in a search to meet the increasing clinical need of early detection and treatment monitoring for AD, with reference to the pathophysiological targets in Alzheimer's brain. These include the pathological aggregations of misfolded proteins such as β-amyloid (Aβ) plagues and neurofibrillary tangles (NFTs), impaired neurotransmitter system, neuroinflammation, as well as deficient synaptic vesicles and glucose utilization. In this article we survey the various PET radiotracers available for AD imaging and discuss their clinical applications especially in terms of early detection and cognitive relevance.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
27
|
Goutal S, Guillermier M, Becker G, Gaudin M, Bramoullé Y, Luxen A, Lemaire C, Plenevaux A, Salmon E, Hantraye P, Barret O, Van Camp N. The pharmacokinetics of [ 18F]UCB-H revisited in the healthy non-human primate brain. EJNMMI Res 2021; 11:36. [PMID: 33826008 PMCID: PMC8026785 DOI: 10.1186/s13550-021-00777-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
Background Positron Emission Tomography (PET) imaging of the Synaptic Vesicle glycoprotein (SV) 2A is a new tool to quantify synaptic density. [18F]UCB-H was one of the first promising SV2A-ligands to be labelled and used in vivo in rodent and human, while limited information on its pharmacokinetic properties is available in the non-human primate. Here, we evaluate the reliability of the three most commonly used modelling approaches for [18F]UCB-H in the non-human cynomolgus primate, adding the coupled fit of the non-displaceable distribution volume (VND) as an alternative approach to improve unstable fit. The results are discussed in the light of the current state of SV2A PET ligands. Results [18F]UCB-H pharmacokinetic data was optimally fitted with a two-compartment model (2TCM), although the model did not always converge (large total volume of distribution (VT) or large uncertainty of the estimate). 2TCM with coupled fit K1/k2 across brain regions stabilized the quantification, and confirmed a lower specific signal of [18F]UCB-H compared to the newest SV2A-ligands. However, the measures of VND and the influx parameter (K1) are similar to what has been reported for other SV2A ligands. These data were reinforced by displacement studies using [19F]UCB-H, demonstrating only 50% displacement of the total [18F]UCB-H signal at maximal occupancy of SV2A. As previously demonstrated in clinical studies, the graphical method of Logan provided a more robust estimate of VT with only a small bias compared to 2TCM. Conclusions Modeling issues with a 2TCM due to a slow component have previously been reported for other SV2A ligands with low specific binding, or after blocking of specific binding. As all SV2A ligands share chemical structural similarities, we hypothesize that this slow binding component is common for all SV2A ligands, but only hampers quantification when specific binding is low. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00777-8.
Collapse
Affiliation(s)
- Sébastien Goutal
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Martine Guillermier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Guillaume Becker
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Mylène Gaudin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Yann Bramoullé
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - André Luxen
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Christian Lemaire
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Alain Plenevaux
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Eric Salmon
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liege, Allee du 6 Aout, 8, Sart Tilman B30, 4000, Liege, Belgium
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Olivier Barret
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire Des Maladies Neurodégénératives, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France.
| |
Collapse
|
28
|
Thomsen MB, Jacobsen J, Lillethorup TP, Schacht AC, Simonsen M, Romero-Ramos M, Brooks DJ, Landau AM. In vivo imaging of synaptic SV2A protein density in healthy and striatal-lesioned rats with [11C]UCB-J PET. J Cereb Blood Flow Metab 2021; 41:819-830. [PMID: 32538280 PMCID: PMC7983510 DOI: 10.1177/0271678x20931140] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 12/30/2022]
Abstract
The number of functionally active synapses provides a measure of neural integrity, with reductions observed in neurodegenerative disorders. [11C]UCB-J binds to synaptic vesicle 2A (SV2A) transmembrane protein located in secretory vesicles. We aimed to assess [11C]UCB-J PET as an in vivo biomarker of regional cerebral synaptic SV2A density in rat lesion models of neurodegeneration. Healthy anesthetized rats had [11C]UCB-J PET and arterial blood sampling. We compared different models describing [11C]UCB-J brain uptake kinetics to determine its regional distribution. Blocking studies were performed with levetiracetam (LEV), an antiepileptic SV2A antagonist. Tracer binding was measured in rodent unilateral acute lesion models of Parkinsonism and Huntington's disease, induced with 6-hydroxydopamine (6-OHDA) and quinolinic acid (QA), respectively. [3H]UCB-J autoradiography was performed in postmortem tissue. Rat brain showed high and fast [11C]UCB-J uptake and washout with up to 80% blockade by LEV. [11C]UCB-J PET showed a 6.2% decrease in ipsilateral striatal SV2A binding after 6-OHDA and 39.3% and 55.1% decreases after moderate and high dose QA confirmed by autoradiography. In conclusion, [11C]UCB-J PET provides a good in vivo marker of synaptic SV2A density which can potentially be followed longitudinally along with synaptic responses to putative neuroprotective agents in models of neurodegeneration.
Collapse
Affiliation(s)
- Majken B Thomsen
- Department of Nuclear Medicine and PET Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jan Jacobsen
- Department of Nuclear Medicine and PET Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thea P Lillethorup
- Department of Nuclear Medicine and PET Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anna C Schacht
- Department of Nuclear Medicine and PET Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mette Simonsen
- Department of Nuclear Medicine and PET Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - David J Brooks
- Department of Nuclear Medicine and PET Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Translational and Clinical Research Institute, Newcastle upon Tyne University, Newcastle upon Tyne, UK
| | - Anne M Landau
- Department of Nuclear Medicine and PET Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
29
|
Wenzel M, Leunig A, Han S, Peterka DS, Yuste R. Prolonged anesthesia alters brain synaptic architecture. Proc Natl Acad Sci U S A 2021; 118:e2023676118. [PMID: 33568534 PMCID: PMC7924219 DOI: 10.1073/pnas.2023676118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Prolonged medically induced coma (pMIC) is carried out routinely in intensive care medicine. pMIC leads to cognitive impairment, yet the underlying neuromorphological correlates are still unknown, as no direct studies of MIC exceeding ∼6 h on neural circuits exist. Here, we establish pMIC (up to 24 h) in adolescent and mature mice, and combine longitudinal two-photon imaging of cortical synapses with repeated behavioral object recognition assessments. We find that pMIC affects object recognition, and that it is associated with enhanced synaptic turnover, generated by enhanced synapse formation during pMIC, while the postanesthetic period is dominated by synaptic loss. Our results demonstrate major side effects of prolonged anesthesia on neural circuit structure.
Collapse
Affiliation(s)
- Michael Wenzel
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Alexander Leunig
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Shuting Han
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Darcy S Peterka
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Rafael Yuste
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY 10027
| |
Collapse
|
30
|
Bagnato F, Gauthier SA, Laule C, Moore GRW, Bove R, Cai Z, Cohen-Adad J, Harrison DM, Klawiter EC, Morrow SA, Öz G, Rooney WD, Smith SA, Calabresi PA, Henry RG, Oh J, Ontaneda D, Pelletier D, Reich DS, Shinohara RT, Sicotte NL. Imaging Mechanisms of Disease Progression in Multiple Sclerosis: Beyond Brain Atrophy. J Neuroimaging 2021; 30:251-266. [PMID: 32418324 DOI: 10.1111/jon.12700] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
Clinicians involved with different aspects of the care of persons with multiple sclerosis (MS) and scientists with expertise on clinical and imaging techniques convened in Dallas, TX, USA on February 27, 2019 at a North American Imaging in Multiple Sclerosis Cooperative workshop meeting. The aim of the workshop was to discuss cardinal pathobiological mechanisms implicated in the progression of MS and novel imaging techniques, beyond brain atrophy, to unravel these pathologies. Indeed, although brain volume assessment demonstrates changes linked to disease progression, identifying the biological mechanisms leading up to that volume loss are key for understanding disease mechanisms. To this end, the workshop focused on the application of advanced magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging techniques to assess and measure disease progression in both the brain and the spinal cord. Clinical translation of quantitative MRI was recognized as of vital importance, although the need to maintain a relatively short acquisition time mandated by most radiology departments remains the major obstacle toward this effort. Regarding PET, the panel agreed upon its utility to identify ongoing pathological processes. However, due to costs, required expertise, and the use of ionizing radiation, PET was not considered to be a viable option for ongoing care of persons with MS. Collaborative efforts fostering robust study designs and imaging technique standardization across scanners and centers are needed to unravel disease mechanisms leading to progression and discovering medications halting neurodegeneration and/or promoting repair.
Collapse
Affiliation(s)
- Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Susan A Gauthier
- Judith Jaffe Multiple Sclerosis Center, Department of Neurology, Feil Family Brain and Mind Institute, and Department of Radiology, Weill Cornell Medicine, New York, NY
| | - Cornelia Laule
- Department of Radiology, Pathology, and Laboratory Medicine, Department of Physics and Astronomy, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - George R Wayne Moore
- Department of Pathology and Laboratory Medicine, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Riley Bove
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University, New Haven, CT
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal and Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Quebec, Canada
| | - Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD
| | - Eric C Klawiter
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sarah A Morrow
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Gülin Öz
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN
| | - William D Rooney
- Advanced Imaging Research Center, Departments of Biomedical Engineering, Neurology, and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | - Seth A Smith
- Radiology and Radiological Sciences and Vanderbilt University Imaging Institute, Vanderbilt University Medical Center, and Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Roland G Henry
- Departments of Neurology, Radiology and Biomedical Imaging, and the UC San Francisco & Berkeley Bioengineering Graduate Group, University of California San Francisco, San Francisco, CA
| | - Jiwon Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Neurology, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Daniel Pelletier
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
| | -
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
31
|
Abstract
Neuroimaging with positron emission tomography (PET) is the most powerful tool for understanding pharmacology, neurochemistry, and pathology in the living human brain. This technology combines high-resolution scanners to measure radioactivity throughout the human body with specific, targeted radioactive molecules, which allow measurements of a myriad of biological processes in vivo. While PET brain imaging has been active for almost 40 years, the pace of development for neuroimaging tools, known as radiotracers, and for quantitative analytical techniques has increased dramatically over the past decade. Accordingly, the fundamental questions that can be addressed with PET have expanded in basic neurobiology, psychiatry, neurology, and related therapeutic development. In this review, we introduce the field of human PET neuroimaging, some of its conceptual underpinnings, and motivating questions. We highlight some of the more recent advances in radiotracer development, quantitative modeling, and applications of PET to the study of the human brain.
Collapse
Affiliation(s)
- Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA;
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
32
|
Becker G, Dammicco S, Bahri MA, Salmon E. The Rise of Synaptic Density PET Imaging. Molecules 2020; 25:molecules25102303. [PMID: 32422902 PMCID: PMC7288098 DOI: 10.3390/molecules25102303] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 11/16/2022] Open
Abstract
Many neurological disorders are related to synaptic loss or pathologies. Before the boom of positrons emission tomography (PET) imaging of synapses, synaptic quantification could only be achieved in vitro on brain samples after autopsy or surgical resections. Until the mid-2010s, electron microscopy and immunohistochemical labelling of synaptic proteins were the gold-standard methods for such analyses. Over the last decade, several PET radiotracers for the synaptic vesicle 2A protein have been developed to achieve in vivo synapses visualization and quantification. Different strategies were used, namely radiolabelling with either 11C or 18F, preclinical development in rodent and non-human primates, and binding quantification with different kinetic modelling methods. This review provides an overview of these PET tracers and underlines their perspectives and limitations by focusing on radiochemical aspects, as well as preclinical proof-of-concept and the main clinical outcomes described so far.
Collapse
|
33
|
Nazeri A, Schifani C, Anderson JAE, Ameis SH, Voineskos AN. In Vivo Imaging of Gray Matter Microstructure in Major Psychiatric Disorders: Opportunities for Clinical Translation. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 5:855-864. [PMID: 32381477 DOI: 10.1016/j.bpsc.2020.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022]
Abstract
Postmortem studies reveal that individuals with major neuropsychiatric disorders such as schizophrenia and autism spectrum disorder have gray matter microstructural abnormalities. These include abnormalities in neuropil organization, expression of proteins supporting neuritic and synaptic integrity, and myelination. Genetic and postmortem studies suggest that these changes may be causally linked to the pathogenesis of these disorders. Advances in diffusion-weighted magnetic resonance image (dMRI) acquisition techniques and biophysical modeling allow for the quantification of gray matter microstructure in vivo. While several biophysical models for imaging microstructural properties are available, one in particular, neurite orientation dispersion and density imaging (NODDI), holds great promise for clinical applications. NODDI can be applied to both gray and white matter and requires only a single extra shell beyond a standard dMRI acquisition. Since its development only a few years ago, the NODDI algorithm has been used to characterize gray matter microstructure in schizophrenia, Alzheimer's disease, healthy aging, and development. These investigations have shown that microstructural findings in vivo, using NODDI, align with postmortem findings. Not only do NODDI and other advanced dMRI-based modeling methods provide a window into the brain previously only available postmortem, but they may be more sensitive to certain brain changes than conventional magnetic resonance imaging approaches. This opens up exciting new possibilities for clinicians to more rapidly detect disease signatures and allows earlier intervention in the course of the disease. Given that neurites and gray matter microstructure have the capacity to rapidly remodel, these novel dMRI-based methods represent an opportunity to noninvasively monitor neuroplastic changes posttherapy within much shorter time scales.
Collapse
Affiliation(s)
- Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Christin Schifani
- Kimel Family Translational Imaging Genetics Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - John A E Anderson
- Kimel Family Translational Imaging Genetics Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Stephanie H Ameis
- Margaret and Wallace McCain Centre for Child, Youth and Family Mental Health, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Centre for Brain and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Aristotle N Voineskos
- Kimel Family Translational Imaging Genetics Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
34
|
McCluskey SP, Plisson C, Rabiner EA, Howes O. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. Eur J Nucl Med Mol Imaging 2020; 47:451-489. [PMID: 31541283 PMCID: PMC6974496 DOI: 10.1007/s00259-019-04488-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE A limit on developing new treatments for a number of central nervous system (CNS) disorders has been the inadequate understanding of the in vivo pathophysiology underlying neurological and psychiatric disorders and the lack of in vivo tools to determine brain penetrance, target engagement, and relevant molecular activity of novel drugs. Molecular neuroimaging provides the tools to address this. This article aims to provide a state-of-the-art review of new PET tracers for CNS targets, focusing on developments in the last 5 years for targets recently available for in-human imaging. METHODS We provide an overview of the criteria used to evaluate PET tracers. We then used the National Institute of Mental Health Research Priorities list to identify the key CNS targets. We conducted a PubMed search (search period 1st of January 2013 to 31st of December 2018), which yielded 40 new PET tracers across 16 CNS targets which met our selectivity criteria. For each tracer, we summarised the evidence of its properties and potential for use in studies of CNS pathophysiology and drug evaluation, including its target selectivity and affinity, inter and intra-subject variability, and pharmacokinetic parameters. We also consider its potential limitations and missing characterisation data, but not specific applications in drug development. Where multiple tracers were present for a target, we provide a comparison of their properties. RESULTS AND CONCLUSIONS Our review shows that multiple new tracers have been developed for proteinopathy targets, particularly tau, as well as the purinoceptor P2X7, phosphodiesterase enzyme PDE10A, and synaptic vesicle glycoprotein 2A (SV2A), amongst others. Some of the most promising of these include 18F-MK-6240 for tau imaging, 11C-UCB-J for imaging SV2A, 11C-CURB and 11C-MK-3168 for characterisation of fatty acid amide hydrolase, 18F-FIMX for metabotropic glutamate receptor 1, and 18F-MNI-444 for imaging adenosine 2A. Our review also identifies recurrent issues within the field. Many of the tracers discussed lack in vivo blocking data, reducing confidence in selectivity. Additionally, late-stage identification of substantial off-target sites for multiple tracers highlights incomplete pre-clinical characterisation prior to translation, as well as human disease state studies carried out without confirmation of test-retest reproducibility.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| | - Christophe Plisson
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
35
|
Cybulska K, Perk L, Booij J, Laverman P, Rijpkema M. Huntington's Disease: A Review of the Known PET Imaging Biomarkers and Targeting Radiotracers. Molecules 2020; 25:molecules25030482. [PMID: 31979301 PMCID: PMC7038198 DOI: 10.3390/molecules25030482] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/19/2022] Open
Abstract
Huntington’s disease (HD) is a fatal neurodegenerative disease caused by a CAG expansion mutation in the huntingtin gene. As a result, intranuclear inclusions of mutant huntingtin protein are formed, which damage striatal medium spiny neurons (MSNs). A review of Positron Emission Tomography (PET) studies relating to HD was performed, including clinical and preclinical data. PET is a powerful tool for visualisation of the HD pathology by non-invasive imaging of specific radiopharmaceuticals, which provide a detailed molecular snapshot of complex mechanistic pathways within the brain. Nowadays, radiochemists are equipped with an impressive arsenal of radioligands to accurately recognise particular receptors of interest. These include key biomarkers of HD: adenosine, cannabinoid, dopaminergic and glutamateric receptors, microglial activation, phosphodiesterase 10 A and synaptic vesicle proteins. This review aims to provide a radiochemical picture of the recent developments in the field of HD PET, with significant attention devoted to radiosynthetic routes towards the tracers relevant to this disease.
Collapse
Affiliation(s)
- Klaudia Cybulska
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 EZ Nijmegen, The Netherlands; (J.B.); (P.L.); (M.R.)
- Radboud Translational Medicine B.V., Radboud University Medical Center, Geert Grooteplein 21 (route 142), 6525 EZ Nijmegen, The Netherlands;
- Correspondence:
| | - Lars Perk
- Radboud Translational Medicine B.V., Radboud University Medical Center, Geert Grooteplein 21 (route 142), 6525 EZ Nijmegen, The Netherlands;
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 EZ Nijmegen, The Netherlands; (J.B.); (P.L.); (M.R.)
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Peter Laverman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 EZ Nijmegen, The Netherlands; (J.B.); (P.L.); (M.R.)
| | - Mark Rijpkema
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 EZ Nijmegen, The Netherlands; (J.B.); (P.L.); (M.R.)
| |
Collapse
|
36
|
Rokka J, Schlein E, Eriksson J. Improved synthesis of SV2A targeting radiotracer [ 11C]UCB-J. EJNMMI Radiopharm Chem 2019; 4:30. [PMID: 31784919 PMCID: PMC6884603 DOI: 10.1186/s41181-019-0080-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/18/2019] [Indexed: 12/30/2022] Open
Abstract
Introduction [11C]UCB-J is a tracer developed for PET (positron emission tomography) that has high affinity towards synaptic vesicle glycoprotein 2A (SV2A), a protein believed to participate in the regulation of neurotransmitter release in neurons and endocrine cells. The localisation of SV2A in the synaptic terminals makes it a viable target for in vivo imaging of synaptic density in the brain. Several SV2A targeting compounds have been evaluated as PET tracers, including [11C]UCB-J, with the aim to facilitate studies of synaptic density in neurological diseases. The original two-step synthesis method failed in our hands to produce sufficient amounts of [11C]UCB-J, but served as an excellent starting point for further optimizations towards a high yielding and simplified one-step method. [11C]Methyl iodide was trapped in a clear THF-water solution containing the trifluoroborate substituted precursor, potassium carbonate and palladium complex. The resulting reaction mixture was heated at 70 °C for 4 min to produce [11C]UCB-J. Results After semi-preparative HPLC purification and reformulation in 10% ethanol/phosphate buffered saline, the product was obtained in 39 ± 5% radiochemical yield based on [11C]methyl iodide, corresponding to 1.8 ± 0.5 GBq at EOS. The radiochemical purity was > 99% and the molar activity was 390 ± 180 GBq/μmol at EOS. The product solution contained < 2 ppb palladium. Conclusions A robust and high yielding production method has been developed for [11C]UCB-J, suitable for both preclinical and clinical PET applications.
Collapse
Affiliation(s)
- Johanna Rokka
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Rudbeck Laboratory, 751 85, Uppsala, Sweden
| | - Eva Schlein
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Rudbeck Laboratory, 751 85, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Uppsala Biomedical Center, Uppsala University, SE-751 23, Uppsala, Sweden. .,PET Centre, Uppsala University Hospital, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
37
|
AD molecular: Molecular imaging of Alzheimer's disease: PET imaging of neurotransmitter systems. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019. [PMID: 31481161 DOI: 10.1016/bs.pmbts.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Current understanding of Alzheimer's disease (AD) pathogenesis relies on the observed accumulations of amyloid β and phosphorylated tau aggregates that are thought to play key roles in initiating or propagating disease. However, other processes including changes in synaptic proteins and neurotransmitter loss have been suggested as important etiologies or contributors. Positron emission tomography (PET) imaging allows in vivo investigations of molecular changes associated with AD. PET imaging with multiple radiotracers can be used in combination with other modalities such as magnetic resonance imaging (MRI), and with assessments of cognition and neuropsychiatric symptoms to investigate the molecular underpinnings of AD. Studies of synaptic protein changes may improve the understanding of disease mechanisms and provide valuable markers of disease progression and therapeutic efficacy. This chapter will illustrate the importance of in vivo molecular imaging in the study of AD with a specific emphasis on PET and radioligands for several non-amyloid targets.
Collapse
|
38
|
Synthesis and in vivo evaluation of [ 18F]UCB-J for PET imaging of synaptic vesicle glycoprotein 2A (SV2A). Eur J Nucl Med Mol Imaging 2019; 46:1952-1965. [PMID: 31175396 DOI: 10.1007/s00259-019-04357-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/02/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE Synaptic abnormalities have been implicated in a variety of neuropsychiatric disorders, including epilepsy, Alzheimer's disease, and schizophrenia. Hence, PET imaging of the synaptic vesicle glycoprotein 2A (SV2A) may be a valuable in vivo biomarker for neurologic and psychiatric diseases. We previously developed [11C]UCB-J, a PET radiotracer with high affinity and selectivity toward SV2A; however, the short radioactive half-life (20 min for 11C) places some limitations on its broader application. Herein, we report the first synthesis of the longer-lived 18F-labeled counterpart (half-life: 110 min), [18F]UCB-J, and its evaluation in nonhuman primates. METHODS [18F]UCB-J was synthesized from the iodonium precursors. PET imaging experiments with [18F]UCB-J were conducted in rhesus monkeys to assess the pharmacokinetic and in vivo binding properties. Arterial samples were taken for analysis of radioactive metabolites and generation of input functions. Regional time-activity curves were analyzed using the one-tissue compartment model to derive regional distribution volumes and binding potentials for comparison with [11C]UCB-J. RESULTS [18F]UCB-J was prepared in high radiochemical and enantiomeric purity, but low radiochemical yield. Evaluation in nonhuman primates indicated that the radiotracer displayed pharmacokinetic and imaging characteristics similar to those of [11C]UCB-J, with moderate metabolism rate, high brain uptake, fast and reversible binding kinetics, and high specific binding signals. CONCLUSION We have accomplished the first synthesis of the novel SV2A radiotracer [18F]UCB-J. [18F]UCB-J is demonstrated to be an excellent imaging agent and may prove to be useful for imaging and quantification of SV2A expression, and synaptic density, in humans.
Collapse
|
39
|
Finnema SJ, Rossano S, Naganawa M, Henry S, Gao H, Pracitto R, Maguire RP, Mercier J, Kervyn S, Nicolas J, Klitgaard H, DeBruyn S, Otoul C, Martin P, Muglia P, Matuskey D, Nabulsi NB, Huang Y, Kaminski RM, Hannestad J, Stockis A, Carson RE. A single-center, open-label positron emission tomography study to evaluate brivaracetam and levetiracetam synaptic vesicle glycoprotein 2A binding in healthy volunteers. Epilepsia 2019; 60:958-967. [PMID: 30924924 PMCID: PMC6532410 DOI: 10.1111/epi.14701] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Brivaracetam (BRV) and levetiracetam (LEV) are antiepileptic drugs that bind synaptic vesicle glycoprotein 2A (SV2A). In vitro and in vivo animal studies suggest faster brain penetration and SV2A occupancy (SO) after dosing with BRV than LEV. We evaluated human brain penetration and SO time course of BRV and LEV at therapeutically relevant doses using the SV2A positron emission tomography (PET) tracer 11 C-UCB-J (EP0074; NCT02602860). METHODS Healthy volunteers were recruited into three cohorts. Cohort 1 (n = 4) was examined with PET at baseline and during displacement after intravenous BRV (100 mg) or LEV (1500 mg). Cohort 2 (n = 5) was studied during displacement and 4 hours postdose (BRV 50-200 mg or LEV 1500 mg). Cohort 3 (n = 4) was examined at baseline and steady state after 4 days of twice-daily oral dosing of BRV (50-100 mg) and 4 hours postdose of LEV (250-600 mg). Half-time of 11 C-UCB-J signal change was computed from displacement measurements. Half-saturation concentrations (IC50 ) were determined from calculated SO. RESULTS Observed tracer displacement half-times were 18 ± 6 minutes for BRV (100 mg, n = 4), 9.7 and 10.1 minutes for BRV (200 mg, n = 2), and 28 ± 6 minutes for LEV (1500 mg, n = 6). Estimated corrected half-times were 8 minutes shorter. The SO was 66%-70% for 100 mg intravenous BRV, 84%-85% for 200 mg intravenous BRV, and 78%-84% for intravenous 1500 mg LEV. The IC50 of BRV (0.46 μg/mL) was 8.7-fold lower than of LEV (4.02 μg/mL). BRV data fitted a single SO versus plasma concentration relationship. Steady state SO for 100 mg BRV was 86%-87% (peak) and 76%-82% (trough). SIGNIFICANCE BRV achieves high SO more rapidly than LEV when intravenously administered at therapeutic doses. Thus, BRV may have utility in treating acute seizures; further clinical studies are needed for confirmation.
Collapse
Affiliation(s)
- Sjoerd J. Finnema
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | - Samantha Rossano
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
- Department of Biomedical EngineeringYale UniversityNew HavenConnecticut
| | - Mika Naganawa
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | - Shannan Henry
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | - Hong Gao
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | - Richard Pracitto
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | | | | | | | | | | | | | | | | | | | - David Matuskey
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | - Nabeel B. Nabulsi
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | - Yiyun Huang
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
| | | | | | | | - Richard E. Carson
- Department of Radiology and Biomedical ImagingPositron Emission Tomography CenterYale UniversityNew HavenConnecticut
- Department of Biomedical EngineeringYale UniversityNew HavenConnecticut
| |
Collapse
|
40
|
Li S, Cai Z, Wu X, Holden D, Pracitto R, Kapinos M, Gao H, Labaree D, Nabulsi N, Carson RE, Huang Y. Synthesis and in Vivo Evaluation of a Novel PET Radiotracer for Imaging of Synaptic Vesicle Glycoprotein 2A (SV2A) in Nonhuman Primates. ACS Chem Neurosci 2019; 10:1544-1554. [PMID: 30396272 PMCID: PMC6810685 DOI: 10.1021/acschemneuro.8b00526] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Structural disruption and alterations of synapses are associated with many brain disorders including Alzheimer's disease, epilepsy, depression, and schizophrenia. We have previously developed the PET radiotracer 11C-UCB-J for imaging and quantification of synaptic vesicle glycoprotein 2A (SV2A) and synaptic density in nonhuman primates and humans. Here we report the synthesis of a novel radiotracer 18F-SDM-8 and its in vivo evaluation in rhesus monkeys. The in vitro binding assay of SDM-8 showed high SV2A binding affinity ( Ki = 0.58 nM). 18F-SDM-8 was prepared in high molar activity (241.7 MBq/nmol) and radiochemical purity (>98%). In the brain, 18F-SDM-8 displayed very high uptake with peak standardized uptake value (SVU) greater than 8 and fast and reversible kinetics. A displacement study with levetiracetam and blocking studies with UCB-J and levetiracetam demonstrated its binding reversibility and specificity toward SV2A. Regional binding potential values were calculated and ranged from 0.8 in the brainstem to 4.5 in the cingulate cortex. By comparing to 11C-UCB-J, 18F-SDM-8 displayed the same attractive imaging properties: very high brain uptake, appropriate tissue kinetics, and high levels of specific binding. Given the longer half-life of F-18 and the feasibility for central production and multisite distribution, 18F-SDM-8 holds promise as an excellent radiotracer for SV2A and as a biomarker for synaptic density measurement in neurodegenerative diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Xiaoai Wu
- Department of Nuclear Medicine, West China Hospital, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Daniel Holden
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Richard Pracitto
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Michael Kapinos
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Hong Gao
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - David Labaree
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Richard E. Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| |
Collapse
|
41
|
Heurling K, Ashton NJ, Leuzy A, Zimmer ER, Blennow K, Zetterberg H, Eriksson J, Lubberink M, Schöll M. Synaptic vesicle protein 2A as a potential biomarker in synaptopathies. Mol Cell Neurosci 2019; 97:34-42. [PMID: 30796959 DOI: 10.1016/j.mcn.2019.02.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 01/12/2023] Open
Abstract
Measuring synaptic density in vivo using positron emission tomography (PET) imaging-based biomarkers targeting the synaptic vesicle protein 2A (SV2A) has received much attention recently due to its potential research and clinical applications in synaptopathies, including neurodegenerative and psychiatric diseases. Fluid-based biomarkers in proteinopathies have previously been suggested to provide information on pathology and disease status that is complementary to PET-based measures, and the same can be hypothesized with respect to SV2A. This review provides an overview of the current state of SV2A PET imaging as a biomarker of synaptic density, the potential role of fluid-based biomarkers for SV2A, and related future perspectives.
Collapse
Affiliation(s)
- Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South, Maudsley NHS Foundation, London, UK
| | - Antoine Leuzy
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Memory Research Unit, Lund University, Sweden
| | - Eduardo R Zimmer
- Department of Pharmacology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; PET Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Mark Lubberink
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Clinical Memory Research Unit, Lund University, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
42
|
Cai Z, Li S, Matuskey D, Nabulsi N, Huang Y. PET imaging of synaptic density: A new tool for investigation of neuropsychiatric diseases. Neurosci Lett 2019; 691:44-50. [PMID: 30075287 PMCID: PMC6339829 DOI: 10.1016/j.neulet.2018.07.038] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Synaptic vesicle glycoprotein 2A (SV2A) is expressed ubiquitously in neurons of the central nervous system, and is the binding target of the anti-epileptic drug levetiracetam. Because of the availability of positron emission tomography (PET) ligands targeting SV2A, there is increasing enthusiasm on the use of SV2A PET to study a variety of neuropsychiatric diseases. This review discusses the recent development of radioligands for PET imaging of SV2A and their potential use in the research and diagnosis of CNS diseases.
Collapse
Affiliation(s)
- Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA.
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - David Matuskey
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
43
|
Finnema SJ, Nabulsi NB, Eid T, Detyniecki K, Lin SF, Chen MK, Dhaher R, Matuskey D, Baum E, Holden D, Spencer DD, Mercier J, Hannestad J, Huang Y, Carson RE. Imaging synaptic density in the living human brain. Sci Transl Med 2017; 8:348ra96. [PMID: 27440727 DOI: 10.1126/scitranslmed.aaf6667] [Citation(s) in RCA: 355] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/24/2016] [Indexed: 12/26/2022]
Abstract
Chemical synapses are the predominant neuron-to-neuron contact in the central nervous system. Presynaptic boutons of neurons contain hundreds of vesicles filled with neurotransmitters, the diffusible signaling chemicals. Changes in the number of synapses are associated with numerous brain disorders, including Alzheimer's disease and epilepsy. However, all current approaches for measuring synaptic density in humans require brain tissue from autopsy or surgical resection. We report the use of the synaptic vesicle glycoprotein 2A (SV2A) radioligand [(11)C]UCB-J combined with positron emission tomography (PET) to quantify synaptic density in the living human brain. Validation studies in a baboon confirmed that SV2A is an alternative synaptic density marker to synaptophysin. First-in-human PET studies demonstrated that [(11)C]UCB-J had excellent imaging properties. Finally, we confirmed that PET imaging of SV2A was sensitive to synaptic loss in patients with temporal lobe epilepsy. Thus, [(11)C]UCB-J PET imaging is a promising approach for in vivo quantification of synaptic density with several potential applications in diagnosis and therapeutic monitoring of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Sjoerd J Finnema
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA.
| | - Nabeel B Nabulsi
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Tore Eid
- Department of Laboratory Medicine, Yale University, New Haven, CT 06520, USA
| | - Kamil Detyniecki
- Department of Neurology, Yale University, New Haven, CT 06520, USA
| | - Shu-Fei Lin
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Ming-Kai Chen
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Roni Dhaher
- Department of Laboratory Medicine, Yale University, New Haven, CT 06520, USA
| | - David Matuskey
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Evan Baum
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Daniel Holden
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Dennis D Spencer
- Department of Neurosurgery, Yale University, New Haven, CT 06520, USA
| | | | | | - Yiyun Huang
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Richard E Carson
- Yale Positron Emission Tomography Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA. Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
44
|
Schifani C, Hafizi S, Da Silva T, Watts JJ, Khan MS, Mizrahi R. Using molecular imaging to understand early schizophrenia-related psychosis neurochemistry: a review of human studies. Int Rev Psychiatry 2017; 29:555-566. [PMID: 29219634 PMCID: PMC8011813 DOI: 10.1080/09540261.2017.1396205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Schizophrenia is a chronic psychiatric disorder generally preceded by a so-called prodromal phase, which is characterized by attenuated psychotic symptoms. Advances in clinical research have enabled prospective identification of those individuals who are at clinical high risk (CHR) for psychosis, with the power to predict psychosis onset within the near future. Changes in several brain neurochemical systems and molecular mechanisms are implicated in the pathophysiology of schizophrenia and the psychosis spectrum, including the dopaminergic, γ-aminobutyric acid (GABA)-ergic, glutamatergic, endocannabinoid, and immunologic (i.e. glial activation) system and other promising future directions such as synaptic density, which are possible to quantify in vivo using positron emission tomography (PET). This paper aims to review in vivo PET studies in the mentioned systems in the early course of psychosis (i.e. CHR and first-episode psychosis (FEP)). The results of reviewed studies are promising; however, the current understanding of the underlying pathology of psychosis is still limited. Importantly, promising efforts involve the development of novel PET radiotracers targeting systems with growing interest in schizophrenia, like the nociceptive system and synaptic density.
Collapse
Affiliation(s)
- Christin Schifani
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Sina Hafizi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tania Da Silva
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jeremy Joseph Watts
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - M. Saad Khan
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Romina Mizrahi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
45
|
Mercier J, Provins L, Valade A. Discovery and development of SV2A PET tracers: Potential for imaging synaptic density and clinical applications. DRUG DISCOVERY TODAY. TECHNOLOGIES 2017; 25:45-52. [PMID: 29233267 DOI: 10.1016/j.ddtec.2017.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 10/18/2022]
Abstract
Imaging synaptic density in vivo has promise for numerous research and clinical applications in the diagnosis and treatment monitoring of neurodegenerative and psychiatric diseases. Recent developments in the field of PET, such as SV2A human imaging with the novel tracers UCB-A, UCB-H and UCB-J, may help in realizing this potential and bring significant benefit for the patients suffering from these diseases. This review provides an overview of the most recent progress in the field of SV2A PET imaging, its potential for use as a biomarker of synaptic density and the future development areas.
Collapse
|
46
|
Bahri MA, Plenevaux A, Aerts J, Bastin C, Becker G, Mercier J, Valade A, Buchanan T, Mestdagh N, Ledoux D, Seret A, Luxen A, Salmon E. Measuring brain synaptic vesicle protein 2A with positron emission tomography and [ 18F]UCB-H. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:481-486. [PMID: 29124105 PMCID: PMC5671624 DOI: 10.1016/j.trci.2017.08.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction Brain distribution of synaptic vesicle protein 2A was measured with fluorine-18 UCB-H ([18F]UCB-H) and positron emission tomography (PET). Methods Images of synaptic density were acquired in healthy volunteers (two young participants and two seniors). Input function was measured by arterial blood sampling (arterial input function) and derived from PET images using carotid activity (image-derived input function). Logan graphical analysis was used to estimate regional synaptic vesicle protein 2A distribution volume. Results [18F]UCB-H uptake was ubiquitous in cortical and subcortical gray matter. Arterial input function and image-derived input function provided regional distribution volume with a high linear relationship. Discussion The cerebral distribution of [18F]UCB-H is similar to that recently observed with carbon-11 UCB-J ([11C]UCB-J). An accurate [18F]UCB-H quantification can be performed without invasive arterial blood sampling when no suitable reference region is available, using dynamic PET carotid activity. Brain synaptic density can be studied in vivo in normal and pathological aging.
Collapse
Affiliation(s)
| | - Alain Plenevaux
- GIGA-Cyclotron Research Center, University of Liège, Liege, Belgium
| | - Joël Aerts
- GIGA-Cyclotron Research Center, University of Liège, Liege, Belgium
| | - Christine Bastin
- GIGA-Cyclotron Research Center, University of Liège, Liege, Belgium
| | - Guillaume Becker
- GIGA-Cyclotron Research Center, University of Liège, Liege, Belgium
| | | | | | | | | | - Didier Ledoux
- Service des soins intensifs généraux, CHU Liege, Liege, Belgium
| | - Alain Seret
- GIGA-Cyclotron Research Center, University of Liège, Liege, Belgium
| | - André Luxen
- GIGA-Cyclotron Research Center, University of Liège, Liege, Belgium
| | - Eric Salmon
- GIGA-Cyclotron Research Center, University of Liège, Liege, Belgium
| |
Collapse
|
47
|
Becker G, Warnier C, Serrano ME, Bahri MA, Mercier J, Lemaire C, Salmon E, Luxen A, Plenevaux A. Pharmacokinetic Characterization of [ 18F]UCB-H PET Radiopharmaceutical in the Rat Brain. Mol Pharm 2017. [PMID: 28651055 DOI: 10.1021/acs.molpharmaceut.7b00235] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The synaptic vesicle glycoprotein 2A (SV2A), a protein essential to the proper nervous system function, is found in presynaptic vesicles. Thus, SV2A targeting, using dedicated radiotracers combined with positron emission tomography (PET), allows the assessment of synaptic density in the living brain. The first-in-class fluorinated SV2A specific radioligand, [18F]UCB-H, is now available at high activity through an efficient radiosynthesis compliant with current good manufacturing practices (cGMP). We report here a noninvasive method to quantify [18F]UCB-H binding in rat brain with microPET. Validation study in rats confirmed the need of high enantiomeric purity to target SV2A in vivo. We demonstrated the reliability of a population-based input function to quantify SV2A in preclinical microPET setting. Finally, we investigated the in vivo metabolism of [18F]UCB-H and confirmed the negligible amount of radiometabolites in the rat brain. Hence, the in vivo quantification of SV2A using [18F]UCB-H microPET seems a promising tool for the assessment of the synaptic density in the rat brain, and opens the way for longitudinal follow-up in neurodegenerative disease rodent models.
Collapse
Affiliation(s)
- Guillaume Becker
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| | - Corentin Warnier
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| | - Maria Elisa Serrano
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| | - Mohamed Ali Bahri
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| | | | - Christian Lemaire
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| | - Eric Salmon
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| | - André Luxen
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| | - Alain Plenevaux
- GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège , 4000 Liège, Belgium
| |
Collapse
|
48
|
Bao W, Jia H, Finnema S, Cai Z, Carson RE, Huang YH. PET Imaging for Early Detection of Alzheimer's Disease: From Pathologic to Physiologic Biomarkers. PET Clin 2017; 12:329-350. [PMID: 28576171 DOI: 10.1016/j.cpet.2017.03.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This article describes the application of various PET imaging agents in the investigation and diagnosis of Alzheimer's disease (AD), including radiotracers for pathologic biomarkers of AD such as β-amyloid deposits and tau protein aggregates, and the neuroinflammation biomarker 18 kDa translocator protein, as well as physiologic biomarkers, such as cholinergic receptors, glucose metabolism, and the synaptic density biomarker synaptic vesicle glycoprotein 2A. Potential of these biomarkers for early AD diagnosis is also assessed.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, No. 518, East Wuzhong Road, Xuhui District, Shanghai 200235, China
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Beijing 10075, China
| | - Sjoerd Finnema
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA.
| |
Collapse
|
49
|
Danish A, Namasivayam V, Schiedel AC, Müller CE. Interaction of Approved Drugs with Synaptic Vesicle Protein 2A. Arch Pharm (Weinheim) 2017; 350. [PMID: 28220535 DOI: 10.1002/ardp.201700003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/04/2017] [Accepted: 02/05/2017] [Indexed: 11/09/2022]
Abstract
Levetiracetam (LEV) and its recently approved derivative brivaracetam are anti-epileptic drugs with a unique mechanism of action. The synaptic vesicle protein 2A (SV2A) was previously identified as their main target. In the current study, we tested a collection of 500 approved drugs for interaction with the human SV2A protein expressed in Chinese hamster ovary cells. Competition binding studies were performed using cell lysates with high SV2A expression and [3 H]brivaracetam as a radioligand. A hit rate of 3% was obtained, defined as compounds that inhibited radioligand binding by more than 90% at a screening concentration of 20 μM. Subsequent concentration-inhibition curves revealed the antihistaminic prodrug loratadine (Ki = 1.16 μM) and the antimalarial drug quinine (Ki = 2.03 μM) to be the most potent SV2A protein ligands of the investigated drug library. Both compounds were similarly potent as LEV (Ki = 1.74 μM), providing structurally novel scaffolds for SV2A ligands. A pharmacophore model was established, which indicated steric and electronic conformities of brivaracetam with the new SV2A ligands, and preliminary structure-activity relationships were determined. The anti-convulsive effects of the natural product quinine may - at least in part - be explained by interaction with SV2A. Loratadine and quinine represent new lead structures for anti-epileptic drug development.
Collapse
Affiliation(s)
- Azeem Danish
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Vigneshwaran Namasivayam
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Anke C Schiedel
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| |
Collapse
|
50
|
Löscher W, Gillard M, Sands ZA, Kaminski RM, Klitgaard H. Synaptic Vesicle Glycoprotein 2A Ligands in the Treatment of Epilepsy and Beyond. CNS Drugs 2016; 30:1055-1077. [PMID: 27752944 PMCID: PMC5078162 DOI: 10.1007/s40263-016-0384-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The synaptic vesicle glycoprotein SV2A belongs to the major facilitator superfamily (MFS) of transporters and is an integral constituent of synaptic vesicle membranes. SV2A has been demonstrated to be involved in vesicle trafficking and exocytosis, processes crucial for neurotransmission. The anti-seizure drug levetiracetam was the first ligand to target SV2A and displays a broad spectrum of anti-seizure activity in various preclinical models. Several lines of preclinical and clinical evidence, including genetics and protein expression changes, support an important role of SV2A in epilepsy pathophysiology. While the functional consequences of SV2A ligand binding are not fully elucidated, studies suggest that subsequent SV2A conformational changes may contribute to seizure protection. Conversely, the recently discovered negative SV2A modulators, such as UCB0255, counteract the anti-seizure effect of levetiracetam and display procognitive properties in preclinical models. More broadly, dysfunction of SV2A may also be involved in Alzheimer's disease and other types of cognitive impairment, suggesting potential novel therapies for levetiracetam and its congeners. Furthermore, emerging data indicate that there may be important roles for two other SV2 isoforms (SV2B and SV2C) in the pathogenesis of epilepsy, as well as other neurodegenerative diseases. Utilization of recently developed SV2A positron emission tomography ligands will strengthen and reinforce the pharmacological evidence that SV2A is a druggable target, and will provide a better understanding of its role in epilepsy and other neurological diseases, aiding in further defining the full therapeutic potential of SV2A modulation.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| | | | | | | | | |
Collapse
|