1
|
Berckmans L, Schrauwen C, Miranda A, Staelens S, Bertoglio D. Assessing non-invasive quantitative methods for [ 18F]SynVesT-1 PET imaging of synaptic vesicle glycoprotein 2A in the rat brain. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07170-w. [PMID: 40032689 DOI: 10.1007/s00259-025-07170-w] [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: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
PURPOSE Synaptic vesicle glycoprotein 2A (SV2A) is a critical biomarker for evaluating synaptic density in neurological research. Among available radioligands, [18F]SynVesT-1 is increasingly used in PET research because of its extended half-life, while having comparable pharmacokinetic properties to the widely used [11C]UCB-J. However, quantitative application in rat models remains unexplored for [18F]SynVesT-1. This study aims to validate quantitative kinetic modelling methods for [18F]SynVesT-1 and develop non-invasive quantification methods for synaptic density in rats. METHODS First, blood analysis of [18F]SynVesT-1 was performed to generate metabolite-corrected plasma input functions. Then, kinetic modelling was evaluated using compartmental analysis approaches, as well as Logan plot. Furthermore, non-invasive image-derived input functions (IDIF), with and without non-negative matrix factorization (NMF) were compared against the arterial input function (AIF). RESULTS Blood analysis showed that the parent fraction of the tracer decreased over time following a sigmoid curve, while the plasma-to-whole blood ratio remained stable over time (0.89 ± 0.02). The two-tissue compartmental model (2TCM) and Logan plot were determined to be the most accurate methods for quantification of [18F]SynVesT-1 kinetics in rats. Additionally, the results demonstrated strong agreement between AIF-derived and image-derived volume of distribution (VT) values, with both image-derived input approaches (IDIF and IDIF-NMF) performing equally well. CONCLUSION These findings validate kinetic modelling methods for [18F]SynVesT-1 PET, enabling their application in further rat studies for preclinical neuroscience research and prove that image-derived input functions are reliable non-invasive alternatives to AIF.
Collapse
Affiliation(s)
- Lori Berckmans
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium.
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium.
| | - Claudia Schrauwen
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, Antwerp, Belgium
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
- URPhyM-NARILIS, University of Namur, Namur, Belgium
| | - Alan Miranda
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Steven Staelens
- µNeuro Center for Excellence, University of Antwerp, Antwerp, Belgium
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Daniele Bertoglio
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, Antwerp, 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
|
Juengling F, Wuest F, Schirrmacher R, Abele J, Thiel A, Soucy JP, Camicioli R, Garibotto V. PET Imaging in Dementia: Mini-Review and Canadian Perspective for Clinical Use. Can J Neurol Sci 2025; 52:26-38. [PMID: 38433571 DOI: 10.1017/cjn.2024.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
PET imaging is increasingly recognized as an important diagnostic tool to investigate patients with cognitive disturbances of possible neurodegenerative origin. PET with 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), assessing glucose metabolism, provides a measure of neurodegeneration and allows a precise differential diagnosis among the most common neurodegenerative diseases, such as Alzheimer's disease, frontotemporal dementia or dementia with Lewy bodies. PET tracers specific for the pathological deposits characteristic of different neurodegenerative processes, namely amyloid and tau deposits typical of Alzheimer's Disease, allow the visualization of these aggregates in vivo. [18F]FDG and amyloid PET imaging have reached a high level of clinical validity and are since 2022 investigations that can be offered to patients in standard clinical care in most of Canada.This article will briefly review and summarize the current knowledge on these diagnostic tools, their integration into diagnostic algorithms as well as perspectives for future developments.
Collapse
Affiliation(s)
- Freimut Juengling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Faculty, University of Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ralf Schirrmacher
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Jonathan Abele
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Department of Neurology and Neurosurgery, Lady Davis Institute for Medical Research, McGill University, Montréal, QC, Canada
| | - Jean-Paul Soucy
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Valentina Garibotto
- Diagnostic Department, Nuclear Medicine and Molecular Imaging Division, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
Kong Y, Cao L, Xie F, Wang X, Zuo C, Shi K, Rominger A, Huang Q, Xiao J, Jiang D, Guan Y, Ni R. Reduced SV2A and GABA A receptor levels in the brains of type 2 diabetic rats revealed by [ 18F]SDM-8 and [ 18F]flumazenil PET. Biomed Pharmacother 2024; 172:116252. [PMID: 38325265 DOI: 10.1016/j.biopha.2024.116252] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
PURPOSE Type 2 diabetes mellitus (T2DM) is associated with a greater risk of Alzheimer's disease. Synaptic impairment and protein aggregates have been reported in the brains of T2DM models. Here, we assessed whether neurodegenerative changes in synaptic vesicle 2 A (SV2A), γ-aminobutyric acid type A (GABAA) receptor, amyloid-β, tau and receptor for advanced glycosylation end product (RAGE) can be detected in vivo in T2DM rats. METHODS Positron emission tomography (PET) using [18F]SDM-8 (SV2A), [18F]flumazenil (GABAA receptor), [18F]florbetapir (amyloid-β), [18F]PM-PBB3 (tau), and [18F]FPS-ZM1 (RAGE) was carried out in 12-month-old diabetic Zucker diabetic fatty (ZDF) and SpragueDawley (SD) rats. Immunofluorescence staining, Thioflavin S staining, proteomic profiling and pathway analysis were performed on the brain tissues of ZDF and SD rats. RESULTS Reduced cortical [18F]SDM-8 uptake and cortical and hippocampal [18F]flumazenil uptake were observed in 12-month-old ZDF rats compared to SD rats. The regional uptake of [18F]florbetapir and [18F]PM-PBB3 was comparable in the brains of 12-month-old ZDF and SD rats. Immunofluorescence staining revealed Thioflavin S-negative, phospho-tau-positive inclusions in the cortex and hypothalamus in the brains of ZDF rats and the absence of amyloid-beta deposits. The level of GABAA receptors was lower in the cortex of ZDF rats than SD rats. Proteomic analysis further demonstrated that, compared with SD rats, synaptic-related proteins and pathways were downregulated in the hippocampus of ZDF rats. CONCLUSION These findings provide in vivo evidence for regional reductions in SV2A and GABAA receptor levels in the brains of aged T2DM ZDF rats.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Cao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China; Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiuzhe Wang
- Dept. Neurology, Shanghai Sixth People's Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Kuangyu Shi
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Axel Rominger
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianfei Xiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Donglang Jiang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ruiqing Ni
- Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland; Dept. Nuclear Medicine, Inselspital, Bern University Hospital, Bern, Switzerland; Inst. Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Drake LR, Wu Y, Naganawa M, Asch R, Zheng C, Najafzadeh S, Pracitto R, Lindemann M, Li S, Ropchan J, Labaree D, Emery PR, Dias M, Henry S, Nabulsi N, Matuskey D, Hillmer AT, Gallezot JD, Carson RE, Cai Z, Huang Y. First-in-Human Study of 18F-SynVesT-2: An SV2A PET Imaging Probe with Fast Brain Kinetics and High Specific Binding. J Nucl Med 2024; 65:jnumed.123.266470. [PMID: 38360052 PMCID: PMC10924160 DOI: 10.2967/jnumed.123.266470] [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: 07/31/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 02/17/2024] Open
Abstract
PET imaging of synaptic vesicle glycoprotein 2A allows for noninvasive quantification of synapses. This first-in-human study aimed to evaluate the kinetics, test-retest reproducibility, and extent of specific binding of a recently developed synaptic vesicle glycoprotein 2A PET ligand, (R)-4-(3-(18F-fluoro)phenyl)-1-((3-methylpyridin-4-yl)methyl)pyrrolidine-2-one (18F-SynVesT-2), with fast brain kinetics. Methods: Nine healthy volunteers participated in this study and were scanned on a High Resolution Research Tomograph scanner with 18F-SynVesT-2. Five volunteers were scanned twice on 2 different days. Five volunteers were rescanned with preinjected levetiracetam (20 mg/kg, intravenously). Arterial blood was collected to calculate the plasma free fraction and generate the arterial input function. Individual MR images were coregistered to a brain atlas to define regions of interest for generating time-activity curves, which were fitted with 1- and 2-tissue-compartment (1TC and 2TC) models to derive the regional distribution volume (V T). The regional nondisplaceable binding potential (BP ND) was calculated from 1TC V T, using the centrum semiovale (CS) as the reference region. Results: 18F-SynVesT-2 was synthesized with high molar activity (187 ± 69 MBq/nmol, n = 19). The parent fraction of 18F-SynVesT-2 in plasma was 28% ± 8% at 30 min after injection, and the plasma free fraction was high (0.29 ± 0.04). 18F-SynVesT-2 entered the brain quickly, with an SUVpeak of 8 within 10 min after injection. Regional time-activity curves fitted well with both the 1TC and the 2TC models; however, V T was estimated more reliably using the 1TC model. The 1TC V T ranged from 1.9 ± 0.2 mL/cm3 in CS to 7.6 ± 0.8 mL/cm3 in the putamen, with low absolute test-retest variability (6.0% ± 3.6%). Regional BP ND ranged from 1.76 ± 0.21 in the hippocampus to 3.06 ± 0.29 in the putamen. A 20-min scan was sufficient to provide reliable V T and BP ND Conclusion: 18F-SynVesT-2 has fast kinetics, high specific uptake, and low nonspecific uptake in the brain. Consistent with the nonhuman primate results, the kinetics of 18F-SynVesT-2 is faster than the kinetics of 11C-UCB-J and 18F-SynVesT-1 in the human brain and enables a shorter dynamic scan to derive physiologic information on cerebral blood flow and synapse density.
Collapse
Affiliation(s)
| | - Yanjun Wu
- Yale PET Center, New Haven, Connecticut
| | | | - Ruth Asch
- Yale PET Center, New Haven, Connecticut
| | | | | | | | | | - Songye Li
- Yale PET Center, New Haven, Connecticut
| | | | | | | | - Mark Dias
- Yale PET Center, New Haven, Connecticut
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Moallemian S, Salmon E, Bahri MA, Beliy N, Delhaye E, Balteau E, Degueldre C, Phillips C, Bastin C. Multimodal imaging of microstructural cerebral alterations and loss of synaptic density in Alzheimer's disease. Neurobiol Aging 2023; 132:24-35. [PMID: 37717552 DOI: 10.1016/j.neurobiolaging.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 09/19/2023]
Abstract
Multiple neuropathological events are involved in Alzheimer's disease (AD). The current study investigated the concurrence of neurodegeneration, increased iron content, atrophy, and demyelination in AD. Quantitative multiparameter magnetic resonance imaging (MRI) maps providing neuroimaging biomarkers for myelination and iron content along with synaptic density measurements using [18F] UCB-H PET were acquired in 24 AD and 19 Healthy controls (19 males). The whole brain voxel-wise group comparison revealed demyelination in the right hippocampus, while no significant iron content difference was detected. Bilateral atrophy and synaptic density loss were observed in the hippocampus and amygdala. The multivariate GLM (mGLM) analysis shows a bilateral difference in the hippocampus and amygdala, right pallidum, left fusiform and temporal lobe suggesting that these regions are the most affected despite the diverse differences in brain tissue properties in AD. Demyelination was identified as the most affecting factor in the observed differences. Here, the mGLM is introduced as an alternative for multiple comparisons between different modalities, reducing the risk of false positives while informing about the co-occurrence of neuropathological processes in AD.
Collapse
Affiliation(s)
- Soodeh Moallemian
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Eric Salmon
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Mohamed Ali Bahri
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Nikita Beliy
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Emma Delhaye
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Evelyne Balteau
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Christian Degueldre
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Christophe Phillips
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Christine Bastin
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| |
Collapse
|
11
|
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
|
12
|
Ni R. PET imaging in animal models of Parkinson's disease. Behav Brain Res 2023; 438:114174. [PMID: 36283568 DOI: 10.1016/j.bbr.2022.114174] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022]
Abstract
Alpha-synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies and multiple system atrophy, are characterized by aberrant accumulation of alpha-synuclein and synaptic dysfunction leading to motor and cognitive deficits. Animal models of alpha-synucleinopathy have greatly facilitated the mechanistic understanding of the disease and the development of therapeutics. Various transgenic, alpha-synuclein fibril-injected, and toxin-injected animal models of Parkinson's disease and multiple system atrophy that recapitulate the disease pathology have been developed and widely used. Recent advances in positron emission tomography have allowed the noninvasive visualization of molecular alterations, underpinning behavioral dysfunctions in the brains of animal models and the longitudinal monitoring of treatment effects. Imaging studies in these disease animal models have employed multi-tracer PET designs to reveal dopaminergic deficits together with other molecular alterations. This review focuses on the development of new positron emission tomography tracers and studies of alpha-synuclein, synaptic vesicle glycoprotein 2A neurotransmitter receptor deficits such as dopaminergic receptor, dopaminergic transporter, serotonergic receptor, vesicular monoamine transporter 2, hypometabolism, neuroinflammation, mitochondrial dysfunction and leucine rich repeat kinase 2 in animal models of Parkinson's disease. The outstanding challenges and emerging applications are outlined, such as investigating the gut-brain-axis by using positron emission tomography in animal models, and provide a future outlook.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
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
|
14
|
van der Weijden CWJ, Mossel P, Bartels AL, Dierckx RAJO, Luurtsema G, Lammertsma AA, Willemsen ATM, de Vries EFJ. Non-invasive kinetic modelling approaches for quantitative analysis of brain PET studies. Eur J Nucl Med Mol Imaging 2023; 50:1636-1650. [PMID: 36651951 PMCID: PMC10119247 DOI: 10.1007/s00259-022-06057-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/21/2022] [Indexed: 01/19/2023]
Abstract
Pharmacokinetic modelling with arterial sampling is the gold standard for analysing dynamic PET data of the brain. However, the invasive character of arterial sampling prevents its widespread clinical application. Several methods have been developed to avoid arterial sampling, in particular reference region methods. Unfortunately, for some tracers or diseases, no suitable reference region can be defined. For these cases, other potentially non-invasive approaches have been proposed: (1) a population based input function (PBIF), (2) an image derived input function (IDIF), or (3) simultaneous estimation of the input function (SIME). This systematic review aims to assess the correspondence of these non-invasive methods with the gold standard. Studies comparing non-invasive pharmacokinetic modelling methods with the current gold standard methods using an input function derived from arterial blood samples were retrieved from PubMed/MEDLINE (until December 2021). Correlation measurements were extracted from the studies. The search yielded 30 studies that correlated outcome parameters (VT, DVR, or BPND for reversible tracers; Ki or CMRglu for irreversible tracers) from a potentially non-invasive method with those obtained from modelling using an arterial input function. Some studies provided similar results for PBIF, IDIF, and SIME-based methods as for modelling with an arterial input function (R2 = 0.59-1.00, R2 = 0.71-1.00, R2 = 0.56-0.96, respectively), if the non-invasive input curve was calibrated with arterial blood samples. Even when the non-invasive input curve was calibrated with venous blood samples or when no calibration was applied, moderate to good correlations were reported, especially for the IDIF and SIME (R2 = 0.71-1.00 and R2 = 0.36-0.96, respectively). Overall, this systematic review illustrates that non-invasive methods to generate an input function are still in their infancy. Yet, IDIF and SIME performed well, not only with arterial blood calibration, but also with venous or no blood calibration, especially for some tracers without plasma metabolites, which would potentially make these methods better suited for clinical application. However, these methods should still be properly validated for each individual tracer and application before implementation.
Collapse
Affiliation(s)
- Chris W J van der Weijden
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands.,Department of Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands
| | - Pascalle Mossel
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Anna L Bartels
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Antoon T M Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands.
| |
Collapse
|
15
|
van der Weijden CWJ, van der Hoorn A, Wang Y, Willemsen ATM, Dierckx RAJO, Lammertsma AA, de Vries EFJ. Investigation of image-derived input functions for non-invasive quantification of myelin density using [ 11C]MeDAS PET. Neuroimage 2022; 264:119772. [PMID: 36436711 DOI: 10.1016/j.neuroimage.2022.119772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/01/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease. Current treatments are focussed on immune suppression to modulate pathogenic activity that causes myelin damage. New treatment strategies are needed to prevent demyelination and promote remyelination. Development of such myelin repair therapies require a sensitive and specific biomarker for efficacy evaluation. Recently, it has been shown that quantification of myelin density is possible using [11C]MeDAS PET. This method, however, requires arterial blood sampling to generate an arterial input function (AIF). As the invasive nature of arterial sampling will reduce clinical applicability, the purpose of this study was to assess whether an image-derived input function (IDIF) can be used as an alternative way to facilitate its routine clinical use. Six healthy controls and 11 MS patients underwent MRI and [11C]MeDAS PET with arterial blood sampling. The application of both population-based whole blood-to-plasma conversion and metabolite corrections were assessed for the AIF. Next, summed images of the early time frames (0-70 s) and the frame with the highest blood-brain contrast were used to generate IDIFs. IDIFs were created using either the hottest 2, 4, 6 or 12 voxels, or an isocontour of the hottest 10% voxels of the carotid artery. This was followed by blood-to-plasma conversion and metabolite correction of the IDIF. The application of a population-based metabolite correction of the AIF resulted in high correlations of tracer binding (Ki) within subjects, but variable bias across subjects. All IDIFs had a sharper and higher peak in the blood curves than the AIF, most likely due to dispersion during blood sampling. All IDIF methods resulted in similar high correlations within subjects (r = 0.95-0.98), but highly variable bias across subjects (mean slope=0.90-1.09). Therefore, both the use of population based blood-plasma and metabolite corrections and the generation of the image-derived whole-blood curve resulted in substantial bias in [11C]MeDAS PET quantification, due to high inter-subject variability. Consequently, when unbiased quantification of [11C]MeDAS PET data is required, individual AIF needs to be used.
Collapse
Affiliation(s)
- Chris W J van der Weijden
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
| | - Anouk van der Hoorn
- Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
| | - Yanming Wang
- Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Antoon T M Willemsen
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
| | - Rudi A J O Dierckx
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
| | - Adriaan A Lammertsma
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands
| | - Erik F J de Vries
- Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands.
| |
Collapse
|
16
|
Dahl K, Larsson S, Bonn P, Wallin A, Itsenko O, Schöll M. GMP Production of [ 18 F]SynVesT-1, a Radioligand for in vivo PET Imaging of Synaptic Vesicle Glycoprotein 2A. J Labelled Comp Radiopharm 2022; 65:315-322. [PMID: 36044030 DOI: 10.1002/jlcr.4002] [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: 05/30/2022] [Revised: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 11/12/2022]
Abstract
[18 F]SynVesT-1 (also known as [18 F]SDM-8 or [18 F]MNI-1126) is a potent and selective synaptic vesicle glycoprotein 2 (SV2A) positron emission tomography (PET) imaging agent. I order to fulfill the increasing clinical demand of an 18 F-labelled SV2A PET ligand, we have developed a fully automated procedure to provide a sterile and pyrogen-free GMP-compliant product of [18 F]SynVesT-1 suitable for clinical studies in humans. [18 F]SynVesT-1 is synthesized via a rapid copper-mediated radiofluorination protocol. The procedure was developed and established on a commercially available module, TracerMaker (ScanSys Laboratorieteknik ApS, Copenhagen, Denmark), a synthesis platform originally developed to conduct carbon-11 radiochemistry. From ~130 GBq (end-of-bombardment), our newly developed procedure enabled us to prepare [18 F]SynVesT-1 in an isolated radioactivity yield of 14220 ± 800 MBq (n = 3), which corresponds to a radiochemical yield (RCY) of 19.5 ± 0.5%. The radiochemical purity (RCP) and enantiomeric purity of each of the final formulated batches exceeded 98%. The overall synthesis time was 90 min and the molar activity 330 ± 60 GBq/μmol (8.9 ± 1.6 Ci/μmol). The produced [18 F]SynVesT-1 was stable over 8 hours at room temperature and is suitable for in vivo PET imaging studies in human subjects.
Collapse
Affiliation(s)
- Kenneth Dahl
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Stefan Larsson
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Peter Bonn
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anita Wallin
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Oleksiy Itsenko
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Michael Schöll
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Dementia Research Centre, Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
17
|
Spatio-Temporal Alterations in Synaptic Density During Epileptogenesis in the Rat Brain. Neuroscience 2022; 499:142-151. [PMID: 35878719 DOI: 10.1016/j.neuroscience.2022.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/07/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022]
Abstract
Synaptic vesicle glycoprotein 2A (SV2A) is a transmembrane protein that binds levetiracetam and is involved in neurotransmission via an unknown mechanism. SV2A-immunoreactivity is reduced in animal models of epilepsy, and in postmortem hippocampi from patients with temporal lobe epilepsy. It is not known if other regions outside the hippocampus are affected in epilepsy, and whether SV2A is expression permanently reduced or regulated over time. In this study, we induced a generalized status epilepticus (SE) by systemic administration of lithium-pilocarpine to adult female rats. The brains from all animals experiencing SE were collected at different time points after the treatment. The radiotracer, [11C]-UCB-J, binds to SV2A with high affinity, and has been used for in vivo imaging as an a-proxy marker for synaptic density. Here we determined the level of tritiated UCB-J binding by semiquantitative autoradiography in the cerebral cortex, hippocampus, thalamus, and hypothalamus, and in subregions of these. A prominent and highly significant reduction in SV2A binding capacity was observed over the first days after SE in the cerebral cortex and the hippocampus, but not in the thalamus and hypothalamus. The magnitude in reduction was larger and occurred earlier in the hippocampus and the piriform cortex, than in other cortical subregions. Interestingly, in all areas examined, the binding capacity returned to control levels 12 weeks after the SE comparable to the chronic phase. These data show that lithium-pilocarpine-induced epileptogenesis involves both loss and gain of synapses in the in a time-dependent manner.
Collapse
|
18
|
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: 53] [Impact Index Per Article: 17.7] [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
|
19
|
Chen B, Marquez-Nostra B, Belitzky E, Toyonaga T, Tong J, Huang Y, Cai Z. PET Imaging in Animal Models of Alzheimer’s Disease. Front Neurosci 2022; 16:872509. [PMID: 35685772 PMCID: PMC9171374 DOI: 10.3389/fnins.2022.872509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The successful development and translation of PET imaging agents targeting β-amyloid plaques and hyperphosphorylated tau tangles have allowed for in vivo detection of these hallmarks of Alzheimer’s disease (AD) antemortem. Amyloid and tau PET have been incorporated into the A/T/N scheme for AD characterization and have become an integral part of ongoing clinical trials to screen patients for enrollment, prove drug action mechanisms, and monitor therapeutic effects. Meanwhile, preclinical PET imaging in animal models of AD can provide supportive information for mechanistic studies. With the recent advancement of gene editing technologies and AD animal model development, preclinical PET imaging in AD models will further facilitate our understanding of AD pathogenesis/progression and the development of novel treatments. In this study, we review the current state-of-the-art in preclinical PET imaging using animal models of AD and suggest future research directions.
Collapse
|
20
|
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
|
21
|
Pazarlar BA, Aripaka SS, Petukhov V, Pinborg L, Khodosevich K, Mikkelsen JD. Expression profile of synaptic vesicle glycoprotein 2A, B, and C paralogues in temporal neocortex tissue from patients with temporal lobe epilepsy (TLE). Mol Brain 2022; 15:45. [PMID: 35578248 PMCID: PMC9109314 DOI: 10.1186/s13041-022-00931-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractSynaptic vesicle glycoprotein-2 (SV2) is a family of proteins consisting of SV2A, SV2B, and SV2C. This protein family has attracted attention in recent years after SV2A was shown to be an epileptic drug target and a perhaps a biomarker of synaptic density. So far, the anatomical localization of these proteins in the rodent and human brain have been reported, but co-expression of SV2 genes on a cellular level, their expressions in the human brain, comparison to radioligand binding, any possible regulation in epilepsy are not known. We have here analyzed the expression of SV2 genes in neuronal subtypes in the temporal neocortex in selected specimens by using single nucleus-RNA sequencing, and performed quantitative PCR in populations of temporal lobe epilepsy (TLE) patients and healthy controls. [3H]-UCB-J autoradiography was performed to analyze the correlation between the mRNA transcript and binding capacity to SV2A. Our data showed that the SV2A transcript is expressed in all glutamatergic and GABAergic cortical subtypes, while SV2B expression is restricted to only the glutamatergic neurons and SV2C has very limited expression in a small subgroup of GABAergic interneurons. The level of [3H]-UCB-J binding and the concentration of SV2A mRNA is strongly correlated in each patient, and the expression is lower in the TLE patients. There is no relationship between SV2A expression and age, sex, seizure frequency, duration of epilepsy, or whether patients were recently treated with levetiracetam or not. Collectively, these findings point out a neuronal subtype-specific distribution of the expression of the three SV2 genes, and the lower levels of both radioligand binding and expression further emphasize the significance of these proteins in this disease.
Collapse
|
22
|
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
|
23
|
Aceves-Serrano L, Neva JL, Doudet DJ. Insight Into the Effects of Clinical Repetitive Transcranial Magnetic Stimulation on the Brain From Positron Emission Tomography and Magnetic Resonance Imaging Studies: A Narrative Review. Front Neurosci 2022; 16:787403. [PMID: 35264923 PMCID: PMC8899094 DOI: 10.3389/fnins.2022.787403] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) has been proposed as a therapeutic tool to alleviate symptoms for neurological and psychiatric diseases such as chronic pain, stroke, Parkinson’s disease, major depressive disorder, and others. Although the therapeutic potential of rTMS has been widely explored, the neurological basis of its effects is still not fully understood. Fortunately, the continuous development of imaging techniques has advanced our understanding of rTMS neurobiological underpinnings on the healthy and diseased brain. The objective of the current work is to summarize relevant findings from positron emission tomography (PET) and magnetic resonance imaging (MRI) techniques evaluating rTMS effects. We included studies that investigated the modulation of neurotransmission (evaluated with PET and magnetic resonance spectroscopy), brain activity (evaluated with PET), resting-state connectivity (evaluated with resting-state functional MRI), and microstructure (diffusion tensor imaging). Overall, results from imaging studies suggest that the effects of rTMS are complex and involve multiple neurotransmission systems, regions, and networks. The effects of stimulation seem to not only be dependent in the frequency used, but also in the participants characteristics such as disease progression. In patient populations, pre-stimulation evaluation was reported to predict responsiveness to stimulation, while post-stimulation neuroimaging measurements showed to be correlated with symptomatic improvement. These studies demonstrate the complexity of rTMS effects and highlight the relevance of imaging techniques.
Collapse
Affiliation(s)
- Lucero Aceves-Serrano
- Department of Medicine/Neurology, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Lucero Aceves-Serrano,
| | - Jason L. Neva
- École de Kinésiologie et des Sciences de l’Activité Physique, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada
| | - Doris J. Doudet
- Department of Medicine/Neurology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
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
|
25
|
Ni R. Positron Emission Tomography in Animal Models of Alzheimer's Disease Amyloidosis: Translational Implications. Pharmaceuticals (Basel) 2021; 14:1179. [PMID: 34832961 PMCID: PMC8623863 DOI: 10.3390/ph14111179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Animal models of Alzheimer's disease amyloidosis that recapitulate cerebral amyloid-beta pathology have been widely used in preclinical research and have greatly enabled the mechanistic understanding of Alzheimer's disease and the development of therapeutics. Comprehensive deep phenotyping of the pathophysiological and biochemical features in these animal models is essential. Recent advances in positron emission tomography have allowed the non-invasive visualization of the alterations in the brain of animal models and in patients with Alzheimer's disease. These tools have facilitated our understanding of disease mechanisms and provided longitudinal monitoring of treatment effects in animal models of Alzheimer's disease amyloidosis. In this review, we focus on recent positron emission tomography studies of cerebral amyloid-beta accumulation, hypoglucose metabolism, synaptic and neurotransmitter receptor deficits (cholinergic and glutamatergic system), blood-brain barrier impairment, and neuroinflammation (microgliosis and astrocytosis) in animal models of Alzheimer's disease amyloidosis. We further propose the emerging targets and tracers for reflecting the pathophysiological changes and discuss outstanding challenges in disease animal models and future outlook in the on-chip characterization of imaging biomarkers towards clinical translation.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
26
|
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
|
27
|
Abstract
The use of PET imaging agents in oncology, cardiovascular disease, and neurodegenerative disease shows the power of this technique in evaluating the molecular and biological characteristics of numerous diseases. These agents provide crucial information for designing therapeutic strategies for individual patients. Novel PET tracers are in continual development and many have potential use in clinical and research settings. This article discusses the potential applications of tracers in diagnostics, the biological characteristics of diseases, the ability to provide prognostic indicators, and using this information to guide treatment strategies including monitoring treatment efficacy in real time to improve outcomes and survival.
Collapse
|
28
|
Salmon E, Bahri MA, Plenevaux A, Becker G, Seret A, Delhaye E, Degueldre C, Balteau E, Lemaire C, Luxen A, Bastin C. In vivo exploration of synaptic projections in frontotemporal dementia. Sci Rep 2021; 11:16092. [PMID: 34373529 PMCID: PMC8352914 DOI: 10.1038/s41598-021-95499-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/20/2021] [Indexed: 11/15/2022] Open
Abstract
The purpose of this exploratory research is to provide data on synaptopathy in the behavioral variant of frontotemporal dementia (bvFTD). Twelve patients with probable bvFTD were compared to 12 control participants and 12 patients with Alzheimer’s disease (AD). Loss of synaptic projections was assessed with [18F]UCBH-PET. Total distribution volume was obtained with Logan method using carotid artery derived input function. Neuroimages were analyzed with SPM12. Verbal fluency, episodic memory and awareness of cognitive impairment were equally impaired in patients groups. Compared to controls, [18F]UCBH uptake tended to decrease in the right anterior parahippocampal gyrus of bvFTD patients. Loss of synaptic projections was observed in the right hippocampus of AD participants, but there was no significant difference in [18F]UCBH brain uptake between patients groups. Anosognosia for clinical disorder was correlated with synaptic density in the caudate nucleus and the anteromedial prefrontal cortex. This study suggests that synaptopathy in bvFTD targets the temporal social brain and self-referential processes.
Collapse
Affiliation(s)
- Eric Salmon
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium.
| | - Mohamed Ali Bahri
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Alain Plenevaux
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Guillaume Becker
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Alain Seret
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Emma Delhaye
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Christian Degueldre
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Evelyne Balteau
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Christian Lemaire
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - André Luxen
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| | - Christine Bastin
- GIGA Cyclotron Research Centre, University of Liège, B30 Sart Tilman, 4000, Liège, Belgium
| |
Collapse
|
29
|
Mertens N, Schmidt ME, Hijzen A, Van Weehaeghe D, Ravenstijn P, Depre M, de Hoon J, Van Laere K, Koole M. Minimally invasive quantification of cerebral P2X7R occupancy using dynamic [ 18F]JNJ-64413739 PET and MRA-driven image derived input function. Sci Rep 2021; 11:16172. [PMID: 34373571 PMCID: PMC8352986 DOI: 10.1038/s41598-021-95715-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 07/29/2021] [Indexed: 01/21/2023] Open
Abstract
[18F]JNJ-64413739 has been evaluated as PET-ligand for in vivo quantification of purinergic receptor subtype 7 receptor (P2X7R) using Logan graphical analysis with a metabolite-corrected arterial plasma input function. In the context of a P2X7R PET dose occupancy study, we evaluated a minimally invasive approach by limiting arterial sampling to baseline conditions. Meanwhile, post dose distribution volumes (VT) under blocking conditions were estimated by combining baseline blood to plasma ratios and metabolite fractions with an MR angiography driven image derived input function (IDIF). Regional postdose VT,IDIF values were compared with corresponding VT,AIF estimates using a arterial input function (AIF), in terms of absolute values, test–retest reliability and receptor occupancy. Compared to an invasive AIF approach, postdose VT,IDIF values and corresponding receptor occupancies showed only limited bias (Bland–Altman analysis: 0.06 ± 0.27 and 3.1% ± 6.4%) while demonstrating a high correlation (Spearman ρ = 0.78 and ρ = 0.98 respectively). In terms of test–retest reliability, regional intraclass correlation coefficients were 0.98 ± 0.02 for VT,IDIF compared to 0.97 ± 0.01 for VT,AIF. These results confirmed that a postdose IDIF, guided by MR angiography and using baseline blood and metabolite data, can be considered for accurate [18F]JNJ-64413739 PET quantification in a repeated PET study design, thus avoiding multiple invasive arterial sampling and increasing dosing flexibility.
Collapse
Affiliation(s)
- Nathalie Mertens
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | | | - Anja Hijzen
- Janssen Research and Development, Beerse, Belgium
| | - Donatienne Van Weehaeghe
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | | | - Marleen Depre
- Center for Clinical Pharmacology, University Hospital and KU Leuven, Leuven, Belgium
| | - Jan de Hoon
- Center for Clinical Pharmacology, University Hospital and KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
30
|
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: 47] [Impact Index Per Article: 11.8] [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
|
31
|
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
|
32
|
Goud NS, Bhattacharya A, Joshi RK, Nagaraj C, Bharath RD, Kumar P. Carbon-11: Radiochemistry and Target-Based PET Molecular Imaging Applications in Oncology, Cardiology, and Neurology. J Med Chem 2021; 64:1223-1259. [PMID: 33499603 DOI: 10.1021/acs.jmedchem.0c01053] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The positron emission tomography (PET) molecular imaging technique has gained its universal value as a remarkable tool for medical diagnosis and biomedical research. Carbon-11 is one of the promising radiotracers that can report target-specific information related to its pharmacology and physiology to understand the disease status. Currently, many of the available carbon-11 (t1/2 = 20.4 min) PET radiotracers are heterocyclic derivatives that have been synthesized using carbon-11 inserted different functional groups obtained from primary and secondary carbon-11 precursors. A spectrum of carbon-11 PET radiotracers has been developed against many of the upregulated and emerging targets for the diagnosis, prognosis, prediction, and therapy in the fields of oncology, cardiology, and neurology. This review focuses on the carbon-11 radiochemistry and various target-specific PET molecular imaging agents used in tumor, heart, brain, and neuroinflammatory disease imaging along with its associated pathology.
Collapse
Affiliation(s)
- Nerella Sridhar Goud
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Ahana Bhattacharya
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Raman Kumar Joshi
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Chandana Nagaraj
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Rose Dawn Bharath
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Pardeep Kumar
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| |
Collapse
|
33
|
Serrano ME, Bahri MA, Becker G, Seret A, Germonpré C, Lemaire C, Giacomelli F, Mievis F, Luxen A, Salmon E, Rogister B, Raedt R, Plenevaux A. Exploring with [ 18F]UCB-H the in vivo Variations in SV2A Expression through the Kainic Acid Rat Model of Temporal Lobe Epilepsy. Mol Imaging Biol 2020; 22:1197-1207. [PMID: 32206990 PMCID: PMC7497718 DOI: 10.1007/s11307-020-01488-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE The main purpose of this study was to understand how the positron emission tomography (PET) measure of the synaptic vesicle 2A (SV2A) protein varies in vivo during the development of temporal lobe epilepsy (TLE) in the kainic acid rat model. PROCEDURES Twenty Sprague Dawley male rats were administered with multiple systemic doses of saline (control group, n = 5) or kainic acid (5 mg/kg/injection, epileptic group, n = 15). Both groups were scanned at the four phases of TLE (early, latent, transition, and chronic phase) with the [18F]UCB-H PET radiotracer and T2-structural magnetic resonance imaging. At the end of the scans (3 months post-status epilepticus), rats were monitored for 7 days with electroencephalography for the detection of spontaneous electrographic seizures. Finally, the immunofluorescence staining for SV2A expression was performed. RESULTS Control rats presented a significant increase in [18F]UCB-H binding at the last two scans, compared with the first ones (p < 0.001). This increase existed but was lower in epileptic animals, producing significant group differences in all the phases of the disease (p < 0.028). Furthermore, the quantification of the SV2A expression in vivo with the [18F]UCB-H radiotracer or ex vivo with immunofluorescence led to equivalent results, with a positive correlation between both. CONCLUSIONS Even if further studies in humans are required, the ability to detect a progressive decrease in SV2A expression during the development of temporal lobe epilepsy supports the use of [18F]UCB-H as a useful tool to differentiate, in vivo, between healthy and epileptic animals along with the development of the epileptic disease.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium.
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK.
| | - Mohamed Ali Bahri
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Guillaume Becker
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
- Radiobiology Unit, SCK•CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Alain Seret
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | | | - Christian Lemaire
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Fabrice Giacomelli
- Nucleis, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Frédéric Mievis
- Nucleis, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - André Luxen
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Eric Salmon
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
- Neurology Department, CHU, Academic Hospital, University of Liège, 4000, Liège, Belgium
| | - Bernard Rogister
- Neurology Department, CHU, Academic Hospital, University of Liège, 4000, Liège, Belgium
- GIGA-Neurosciences, University of Liège, Avenue Hippocrate, 15, 4000, Liège, Belgium
| | | | - Alain Plenevaux
- GIGA, CRC in vivo imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| |
Collapse
|
34
|
Serrano ME, Bahri MA, Becker G, Seret A, Mievis F, Giacomelli F, Lemaire C, Salmon E, Luxen A, Plenevaux A. Quantification of [ 18F]UCB-H Binding in the Rat Brain: From Kinetic Modelling to Standardised Uptake Value. Mol Imaging Biol 2020; 21:888-897. [PMID: 30460626 DOI: 10.1007/s11307-018-1301-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE [18F]UCB-H is a specific positron emission tomography (PET) biomarker for the Synaptic Vesicle protein 2A (SV2A), the binding site of the antiepileptic drug levetiracetam. With a view to optimising acquisition time and simplifying data analysis with this radiotracer, we compared two parameters: the distribution volume (Vt) obtained from Logan graphical analysis using a Population-Based Input Function, and the Standardised Uptake Value (SUV). PROCEDURES Twelve Sprague Dawley male rats, pre-treated with three different doses of levetiracetam were employed to develop the methodology. Three additional kainic acid (KA) treated rats (temporal lobe epilepsy model) were also used to test the procedure. Image analyses focused on: (i) length of the dynamic acquisition (90 versus 60 min); (ii) correlations between Vt and SUV over 20-min consecutive time-frames; (iii) and (iv) evaluation of differences between groups using the Vt and the SUV; and (v) preliminary evaluation of the methodology in the KA epilepsy model. RESULTS A large correlation between the Vt issued from 60 to 90-min acquisitions was observed. Further analyses highlighted a large correlation (r > 0.8) between the Vt and the SUV. Equivalent differences between groups were detected for both parameters, especially in the 20-40 and 40-60-min time-frames. The same results were also obtained with the epilepsy model. CONCLUSIONS Our results enable the acquisition setting to be changed from a 90-min dynamic to a 20-min static PET acquisition. According to a better image quality, the 20-40-min time-frame appears optimal. Due to its equivalence to the Vt, the SUV parameter can be considered in order to quantify [18F]UCB-H uptake in the rat brain. This work, therefore, establishes a starting point for the simplification of SV2A in vivo quantification with [18F]UCB-H, and represents a step forward to the clinical application of this PET radiotracer.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium.
| | - Mohamed Ali Bahri
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Guillaume Becker
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Alain Seret
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Frédéric Mievis
- Nucleis, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Fabrice Giacomelli
- Nucleis, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Christian Lemaire
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Eric Salmon
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - André Luxen
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| | - Alain Plenevaux
- GIGA - CRC In Vivo Imaging, University of Liège, Allée du 6 Août, Building B30, Sart Tilman, 4000, Liège, Belgium
| |
Collapse
|
35
|
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: 23] [Impact Index Per Article: 4.6] [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
|
36
|
Development and In Vivo Preclinical Imaging of Fluorine-18-Labeled Synaptic Vesicle Protein 2A (SV2A) PET Tracers. Mol Imaging Biol 2020; 21:509-518. [PMID: 30084043 DOI: 10.1007/s11307-018-1260-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE Synaptic vesicle protein 2A (SV2A) serves as a biomarker of synaptic density and positron emission tomography (PET) imaging of SV2A could provide a tool to assess progression of neurodegenerative diseases. Two tracers have primarily been reported and characterized in vivo: [11C]UCB-J and [18F]UCB-H. In early human studies, [11C]UCB-J showed promising results, while its F-18-labeled analogue [18F]UCB-H showed suboptimal specific signal in comparison to [11C]UCB-J. Considering the limited use of [11C]UCB-J to facilities with a cyclotron, having a F-18 variant would facilitate large, multicenter imaging trials. We have screened several F-18 derivatives of UCB-J in non-human primates and identified a promising F-18 PET candidate, [18F]MNI-1126, with additional investigations of the racemate [18F]MNI-1038, affording a signal comparable to [11C]UCB-J. PROCEDURES F-18 derivatives of UCB-J and UCB-H were synthesized and administered to non-human primates for microPET imaging. Following screenings, [18F]MNI-1038 (racemate) and [18F]MNI-1126 (R-enantiomer) were identified with the highest signal and favorable kinetics and were selected for further imaging. Kinetic modeling with one- and two-tissue compartmental models, and linear methods were applied to PET data using metabolite-corrected arterial input function. Pre-block scans with levetiracetam (LEV, 10, 30 mg/kg, iv) were performed to determine the tracers' in vivo specificity for SV2A. Two whole-body PET studies were performed with [18F]MNI-1038 in one male and one female rhesus, and radiation absorbed dose estimates and effective dose (ED, ICRP-103) were estimated with OLINDA/EXM 2.0. RESULTS All compounds screened displayed very good brain penetration, with a plasma-free fraction of ~ 40 %. [18F]MNI-1126 and [18F]MNI-1038 showed uptake and distribution the most consistent with UCB-J, while the other derivatives showed suboptimal results, with similar or lower uptake than [18F]UCB-H. VT of [18F]MNI-1126 and [18F]MNI-1038 was high in all gray matter regions (within animal averages ~ 30 ml/cm3) and highly correlated with [11C]UCB-J (r > 0.99). Pre-blocking of [18F]MNI-1126 or [18F]MNI-1038 with LEV showed robust occupancy across all gray matter regions, similar to that reported with [11C]UCB-J (~ 85 % at 30 mg/kg, ~ 65 % at 10 mg/kg). Using the centrum semiovale as a reference region, BPND of [18F]MNI-1126 reached values of up to ~ 30 to 40 % higher than those reported for [11C]UCB-J. From whole-body imaging average ED of [18F]MNI-1038 was estimated to be 22.3 μSv/MBq, with tracer being eliminated via both urinary and hepatobiliary pathways. CONCLUSIONS We have identified a F-18-labeled tracer ([18F]MNI-1126) that exhibits comparable in vivo characteristics and specificity for SV2A to [11C]UCB-J in non-human primates, which makes [18F]MNI-1126 a promising PET radiotracer for imaging SV2A in human trials.
Collapse
|
37
|
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
|
38
|
Cai Z, Li S, Zhang W, Pracitto R, Wu X, Baum E, Finnema SJ, Holden D, Toyonaga T, Lin SF, Lindemann M, Shirali A, Labaree DC, Ropchan J, Nabulsi N, Carson RE, Huang Y. Synthesis and Preclinical Evaluation of an 18F-Labeled Synaptic Vesicle Glycoprotein 2A PET Imaging Probe: [ 18F]SynVesT-2. ACS Chem Neurosci 2020; 11:592-603. [PMID: 31961649 DOI: 10.1021/acschemneuro.9b00618] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Synaptic vesicle glycoprotein 2A (SV2A) is a 12-pass transmembrane glycoprotein ubiquitously expressed in presynaptic vesicles. In vivo imaging of SV2A using PET has potential applications in the diagnosis and prognosis of a variety of neuropsychiatric diseases, e.g., Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, autism, epilepsy, stroke, traumatic brain injury, post-traumatic stress disorder, depression, etc. Herein, we report the synthesis and evaluation of a new 18F-labeled SV2A PET imaging probe, [18F]SynVesT-2, which possesses fast in vivo binding kinetics and high specific binding signals in non-human primate brain.
Collapse
Affiliation(s)
- Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Wenjie Zhang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Richard Pracitto
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Xiaoai Wu
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Evan Baum
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Sjoerd J. Finnema
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Daniel Holden
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Takuya Toyonaga
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Shu-fei Lin
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Marcel Lindemann
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Anupama Shirali
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - David C. Labaree
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Jim Ropchan
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Richard E. Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Stout K, Dunn A, Hoffman C, Miller GW. The Synaptic Vesicle Glycoprotein 2: Structure, Function, and Disease Relevance. ACS Chem Neurosci 2019; 10:3927-3938. [PMID: 31394034 PMCID: PMC11562936 DOI: 10.1021/acschemneuro.9b00351] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The synaptic vesicle glycoprotein 2 (SV2) family is comprised of three paralogues: SV2A, SV2B, and SV2C. In vertebrates, SV2s are 12-transmembrane proteins present on every secretory vesicle, including synaptic vesicles, and are critical to neurotransmission. Structural and functional studies suggest that SV2 proteins may play several roles to promote proper vesicular function. Among these roles are their potential to stabilize the transmitter content of vesicles, to maintain and orient the releasable pool of vesicles, and to regulate vesicular calcium sensitivity to ensure efficient, coordinated release of the transmitter. The SV2 family is highly relevant to human health in a number of ways. First, SV2A plays a role in neuronal excitability and as such is the specific target for the antiepileptic drug levetiracetam. SV2 proteins also act as the target by which potent neurotoxins, particularly botulinum, gain access to neurons and exert their toxicity. Both SV2B and SV2C are increasingly implicated in diseases such as Alzheimer's disease and Parkinson's disease. Interestingly, despite decades of intensive research, their exact function remains elusive. Thus, SV2 proteins are intriguing in their potentially diverse roles within the presynaptic terminal, and several recent developments have enhanced our understanding and appreciation of the protein family. Here, we review the structure and function of SV2 proteins as well as their relevance to disease and therapeutic development.
Collapse
Affiliation(s)
- Kristen Stout
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States
| | - Amy Dunn
- The Jackson Laboratory, Bar Harbor, Maine, United States
| | - Carlie Hoffman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States
| |
Collapse
|
42
|
In vivo imaging of synaptic loss in Alzheimer's disease with [18F]UCB-H positron emission tomography. Eur J Nucl Med Mol Imaging 2019; 47:390-402. [PMID: 31468182 DOI: 10.1007/s00259-019-04461-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/24/2019] [Indexed: 11/27/2022]
Abstract
PURPOSE Loss of brain synapses is an early pathological feature of Alzheimer's disease. The current study assessed synaptic loss in vivo with positron emission tomography and an 18F-labelled radiotracer of the synaptic vesicle protein 2A, [18F]UCB-H. METHODS Twenty-four patients with mild cognitive impairment or Alzheimer's disease and positive [18F]Flutemetamol amyloid-PET were compared to 19 healthy controls. [18F]UCB-H brain uptake was quantified with Logan graphical analysis using an image-derived blood input function. SPM12 and regions-of-interest (ROI) analyses were used for group comparisons of regional brain distribution volumes and for correlation with cognitive measures. RESULTS A significant decrease of [18F]UCB-H uptake was observed in several cortical areas (11 to 18% difference) and in the thalamus (16% difference), with the largest effect size in the hippocampus (31% difference). Reduced hippocampal uptake was related to patients' cognitive decline (ROI analysis) and unawareness of memory problems (SPM and ROI analyses). CONCLUSIONS The findings thus highlight predominant synaptic loss in the hippocampus, confirming previous autopsy-based studies and a recent PET study with an 11C-labelled SV2A radiotracer. [18F]UCB-H PET allows to image in vivo synaptic changes in Alzheimer's disease and to relate them to patients' cognitive impairment.
Collapse
|
43
|
Perani D, Iaccarino L, Lammertsma AA, Windhorst AD, Edison P, Boellaard R, Hansson O, Nordberg A, Jacobs AH. A new perspective for advanced positron emission tomography-based molecular imaging in neurodegenerative proteinopathies. Alzheimers Dement 2019; 15:1081-1103. [PMID: 31230910 DOI: 10.1016/j.jalz.2019.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
Recent studies in neurodegenerative conditions have increasingly highlighted that the same neuropathology can trigger different clinical phenotypes or, vice-versa, that similar phenotypes can be triggered by different neuropathologies. This evidence has called for the adoption of a pathology spectrum-based approach to study neurodegenerative proteinopathies. These conditions share brain deposition of abnormal protein aggregates, leading to aberrant biochemical, metabolic, functional, and structural changes. Positron emission tomography (PET) is a well-recognized and unique tool for the in vivo assessment of brain neuropathology, and novel PET techniques are emerging for the study of specific protein species. Today, key applications of PET range from early research and clinical diagnostic tools to their use in clinical trials for both participants screening and outcome evaluation. This position article critically reviews the role of distinct PET molecular tracers for different neurodegenerative proteinopathies, highlighting their strengths, weaknesses, and opportunities, with special emphasis on methodological challenges and future applications.
Collapse
Affiliation(s)
- Daniela Perani
- Vita-Salute San Raffaele University, Nuclear Medicine Unit San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Iaccarino
- Vita-Salute San Raffaele University, Nuclear Medicine Unit San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul Edison
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK; Neurology Imaging Unit, Imperial College London, London, UK
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Center for Alzheimer Research, Stockholm, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany; Evangelische Kliniken Bonn gGmbH, Johanniter Krankenhaus, Bonn, Germany.
| | | |
Collapse
|
44
|
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
|
45
|
Evaluating the In Vivo Specificity of [ 18F]UCB-H for the SV2A Protein, Compared with SV2B and SV2C in Rats Using microPET. Molecules 2019; 24:molecules24091705. [PMID: 31052478 PMCID: PMC6538996 DOI: 10.3390/molecules24091705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/28/2019] [Accepted: 04/29/2019] [Indexed: 11/25/2022] Open
Abstract
The synaptic vesicle protein 2 (SV2) is involved in synaptic vesicle trafficking. The SV2A isoform is the most studied and its implication in epilepsy therapy led to the development of the first SV2A PET radiotracer [18F]UCB-H. The objective of this study was to evaluate in vivo, using microPET in rats, the specificity of [18F]UCB-H for SV2 isoform A in comparison with the other two isoforms (B and C) through a blocking assay. Twenty Sprague Dawley rats were pre-treated either with the vehicle, or with specific competitors against SV2A (levetiracetam), SV2B (UCB5203) and SV2C (UCB0949). The distribution volume (Vt, Logan plot, t* 15 min) was obtained with a population-based input function. The Vt analysis for the entire brain showed statistically significant differences between the levetiracetam group and the other groups (p < 0.001), but also between the vehicle and the SV2B group (p < 0.05). An in-depth Vt analysis conducted for eight relevant brain structures confirmed the statistically significant differences between the levetiracetam group and the other groups (p < 0.001) and highlighted the superior and the inferior colliculi along with the cortex as regions also displaying statistically significant differences between the vehicle and SV2B groups (p < 0.05). These results emphasize the in vivo specificity of [18F]UCB-H for SV2A against SV2B and SV2C, confirming that [18F]UCB-H is a suitable radiotracer for in vivo imaging of the SV2A proteins with PET.
Collapse
|
46
|
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: 51] [Impact Index Per Article: 8.5] [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
|
47
|
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: 83] [Impact Index Per Article: 13.8] [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
|
48
|
Abstract
Alzheimer's disease (AD) dementia refers to a particular onset and course of cognitive and functional decline associated with age together with a particular neuropathology. It was first described by Alois Alzheimer in 1906 about a patient whom he first encountered in 1901. Modern clinical diagnostic criteria have been developed, and criteria have also been proposed to recognize preclinical (or presymptomatic) stages of the disease with the use of biomarkers. The primary neuropathology was described by Alzheimer, and in the mid-1980s subsequently evolved into a more specific neuropathologic definition that recognizes the comorbid neuropathologies that frequently contribute to clinical dementia. Alzheimer's disease is now the most common form of neurodegenerative dementia in the United States with a disproportionate disease burden in minority populations. Deficits in the ability to encode and store new memories characterizes the initial stages of the disease. Subsequent progressive changes in cognition and behavior accompany the later stages. Changes in amyloid precursor protein (APP) cleavage and production of the APP fragment beta-amyloid (Aβ) along with hyperphosphorylated tau protein aggregation coalesce to cause reduction in synaptic strength, synaptic loss, and neurodegeneration. Metabolic, vascular, and inflammatory changes, as well as comorbid pathologies are key components of the disease process. Symptomatic treatment offers a modest, clinically measurable effect in cognition, but disease-modifying therapies are desperately needed.
Collapse
Affiliation(s)
- Jose A Soria Lopez
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States; Shiley-Marcos Alzheimer's Disease Research Center, University of California San Diego, La Jolla, CA, United States
| | - Hector M González
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States; Shiley-Marcos Alzheimer's Disease Research Center, University of California San Diego, La Jolla, CA, United States
| | - Gabriel C Léger
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States; Shiley-Marcos Alzheimer's Disease Research Center, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
49
|
Affiliation(s)
- Elizabeth C. Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| | - William J. Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley
| |
Collapse
|
50
|
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
|