1
|
Dahlén AD, Roshanbin S, Aguilar X, Bucher NM, Lopes van den Broek S, Sehlin D, Syvänen S. PET imaging of TREM2 in amyloid-beta induced neuroinflammation. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07358-0. [PMID: 40434494 DOI: 10.1007/s00259-025-07358-0] [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: 02/05/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025]
Abstract
PURPOSE The triggering receptor expressed on myeloid cells 2 (TREM2) has become a promising target for biologics in both monitoring and treating neuroinflammation in Alzheimer's disease (AD). This study aimed to develop and compare bispecific anti-TREM2 antibodies featuring different transferrin receptor (TfR) binders to enhance brain delivery, identifying the most suitable format for in vivo PET imaging of TREM2 in transgenic AD mice. METHODS Three bispecific TREM2-antibody formats were produced and evaluated for their ability to cross the blood-brain barrier (BBB) via TfR-mediated transcytosis and bind TREM2. Blood concentration profiles up to 72 h post-injection (p.i.), and ex vivo brain uptake of iodine-125-labeled antibody constructs were quantified in AppNL-G-F and age-matched wild type (WT) mice using a γ-counter. The best-performing bispecific TREM2-antibody was radiolabeled with iodine-124 and used for in vivo PET imaging of brain TREM2 levels in AppNL-G-F mice at 72 h p.i. Brain TREM2 concentrations were subsequently quantified using ELISA. RESULTS The antibody format carrying two scFv8D3 TfR-binders (IgG-scFv2), demonstrated the highest brain concentrations of all tested bispecific constructs. This antibody also exhibited significantly higher brain concentrations in AppNL-G-F mice compared to WT mice at both 48 and 72 h p.i. This difference was further visualized and quantified through in vivo PET imaging. Moreover, brain concentrations of the antibody ligand correlated with elevated TREM2 levels in brain homogenates. CONCLUSION These findings highlight IgG-scFv2 as a promising radioligand for in vivo PET imaging of TREM2, advancing non-invasive neuroinflammation studies and supporting drug development for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Amelia D Dahlén
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Ximena Aguilar
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Nadja M Bucher
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Sara Lopes van den Broek
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
2
|
Tang S, Harrison DM, Bardhoshi A, Cureton R, Yan X, Parcon PA, Morse CL, Ecker C, Choi S, Pike VW, Innis RB, Zanotti-Fregonara P. Cyclooxygenase-1 and cyclooxygenase-2 densities measured using positron emission tomography are not altered in the brains of individuals with stable multiple sclerosis. J Cereb Blood Flow Metab 2025:271678X251332490. [PMID: 40367389 PMCID: PMC12078256 DOI: 10.1177/0271678x251332490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 05/16/2025]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease affecting the central nervous system that involves immune-mediated demyelination and axonal degeneration. Clinical imaging techniques play a critical role in diagnosing and assessing the prognosis of MS. Magnetic resonance imaging has been most frequently used to visualize demyelination and detect acute and chronic active lesions, which are key indicators of clinical course of illness. Previous research has also highlighted the effectiveness of translocator protein 18-kDa (TSPO) positron emission tomography (PET) imaging for identifying chronic active lesions and progressive pathology. Building on this work, the present study used PET imaging to explore the role of cyclooxygenase-1 and -2 (COX-1 and COX-2)-key enzymes involved in neuroinflammation-in individuals with MS. Five participants with MS were recruited, and lesions were identified using 7 Tesla MRI. No significant differences in COX radioligand binding were observed in the co-registered PET images between lesioned areas and normal-appearing brain tissues, nor between individuals with MS and healthy volunteers. The negative findings underscore the complexity of MS pathology and raise several important considerations for planning future studies using COX PET for imaging in MS.
Collapse
Affiliation(s)
- Shiyu Tang
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Daniel M Harrison
- Dept of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Dept of Neurology, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | - Amanda Bardhoshi
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Raven Cureton
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Xuefeng Yan
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Paul A Parcon
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Christina Ecker
- Dept of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Seongjin Choi
- Dept of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | | |
Collapse
|
3
|
Yan X, Siméon FG, Liow JS, Morse CL, Jana S, Montero Santamaria JA, Jenkins M, Zoghbi SS, Pike VW, Innis RB, Zanotti-Fregonara P. [ 18F]SF51, a novel 18F-labeled PET radioligand for translocator protein 18kDa (TSPO) in brain, works well in monkeys but fails in humans. J Cereb Blood Flow Metab 2025; 45:365-372. [PMID: 39654356 PMCID: PMC11629344 DOI: 10.1177/0271678x241304924] [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: 08/09/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024]
Abstract
[18F]SF51 is a novel radioligand for imaging translocator protein 18 kDa (TSPO) that previously displayed excellent imaging properties in nonhuman primates. This study assessed its performance in human brain and its dosimetry. Seven healthy participants underwent brain PET imaging to measure TSPO binding using a two-tissue compartment model (2TCM) to calculate total distribution volume (VT). This cohort included two high-affinity binders (HABs), three mixed-affinity binders (MABs), and two low-affinity binders (LABs). Two other participants received whole-body scans to assess radiation exposure. Peak brain radioactivity reached a standardized uptake value (SUV) of 1.4 at 3 minutes post-injection, diminishing to 30% of peak by 120 minutes. The average VT for all genotype groups was notably low (<1 mL·cm-3), emphasizing the radioligand's poor binding in brain. [18F]SF51 remained sensitive to the TSPO polymorphism in vivo, as shown by a two-fold difference in VT between HABs and LABs. VT stabilization by 80 minutes post-injection suggested minimal radiometabolite accumulation in brain. The average effective dose was 13.8 ± 0.9 µSv/MBq. Contrary to previously published animal data, [18F]SF51 showed low binding to human TSPO, with uptake remaining influenced by the rs6971 polymorphism. These findings highlight the challenges of developing TSPO radioligands and underscore the significant species differences that may influence translational outcomes.ClinicalTrials.gov identifier: NCT05564429; registered 10/03/2022.
Collapse
Affiliation(s)
- Xuefeng Yan
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Fabrice G Siméon
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Susovan Jana
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Jose A Montero Santamaria
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Madeline Jenkins
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Fu W, Lin Q, Fu Z, Yang T, Shi D, Ma P, Su H, Wang Y, Liu G, Ding J, Shi H, Cheng D. Synthesis and evaluation of TSPO-targeting radioligand [ 18F]F-TFQC for PET neuroimaging in epileptic rats. Acta Pharm Sin B 2025; 15:722-736. [PMID: 40177559 PMCID: PMC11959965 DOI: 10.1016/j.apsb.2024.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/15/2024] [Accepted: 05/25/2024] [Indexed: 04/05/2025] Open
Abstract
The translocator protein (TSPO) positron emission tomography (PET) can noninvasively detect neuroinflammation associated with epileptogenesis and epilepsy. This study explored the role of the TSPO-targeting radioligand [18F]F-TFQC, an m-trifluoromethyl ER176 analog, in the PET neuroimaging of epileptic rats. Initially, [18F]F-TFQC was synthesized with a radiochemical yield of 8%-10% (EOS), a radiochemical purity of over 99%, and a specific activity of 38.21 ± 1.73 MBq/nmol (EOS). After determining that [18F]F-TFQC exhibited good biochemical properties, [18F]F-TFQC PET neuroimaging was performed in epileptic rats at multiple time points in various stages of disease progression. PET imaging showed specific [18F]F-TFQC uptake in the right hippocampus (KA-injected site, i.e., epileptogenic zone), which was most pronounced at 1 week (T/NT 1.63 ± 0.21) and 1 month (T/NT 1.66 ± 0.20). The PET results were further validated using autoradiography and pathological analysis. Thus, [18F]F-TFQC can reflect the TSPO levels and localize the epileptogenic zone, thereby offering the potential for monitoring neuroinflammation and guiding anti-inflammatory treatment in patients with epilepsy.
Collapse
Affiliation(s)
- Wenhui Fu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Qingyu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Zhequan Fu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Tingting Yang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Dai Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Pengcheng Ma
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Hongxing Su
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Yunze Wang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| |
Collapse
|
5
|
Appleton J, Finn Q, Zanotti-Fregonara P, Yu M, Faridar A, Nakawah MO, Zarate C, Carrillo MC, Dickerson BC, Rabinovici GD, Apostolova LG, Masdeu JC, Pascual B. Brain inflammation co-localizes highly with tau in mild cognitive impairment due to early-onset Alzheimer's disease. Brain 2025; 148:119-132. [PMID: 39013020 PMCID: PMC11706285 DOI: 10.1093/brain/awae234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Brain inflammation, with an increased density of microglia and macrophages, is an important component of Alzheimer's disease and a potential therapeutic target. However, it is incompletely characterized, particularly in patients whose disease begins before the age of 65 years and, thus, have few co-pathologies. Inflammation has been usefully imaged with translocator protein (TSPO) PET, but most inflammation PET tracers cannot image subjects with a low-binder TSPO rs6971 genotype. In an important development, participants with any TSPO genotype can be imaged with a novel tracer, 11C-ER176, that has a high binding potential and a more favourable metabolite profile than other TSPO tracers currently available. We applied 11C-ER176 to detect brain inflammation in mild cognitive impairment (MCI) caused by early-onset Alzheimer's disease. Furthermore, we sought to correlate the brain localization of inflammation, volume loss, elevated amyloid-β (Aβ)and tau. We studied brain inflammation in 25 patients with early-onset amnestic MCI (average age 59 ± 4.5 years, 10 female) and 23 healthy controls (average age 65 ± 6.0 years, 12 female), both groups with a similar proportion of all three TSPO-binding affinities. 11C-ER176 total distribution volume (VT), obtained with an arterial input function, was compared across patients and controls using voxel-wise and region-wise analyses. In addition to inflammation PET, most MCI patients had Aβ (n = 23) and tau PET (n = 21). For Aβ and tau tracers, standard uptake value ratios were calculated using cerebellar grey matter as region of reference. Regional correlations among the three tracers were determined. Data were corrected for partial volume effect. Cognitive performance was studied with standard neuropsychological tools. In MCI caused by early-onset Alzheimer's disease, there was inflammation in the default network, reaching statistical significance in precuneus and lateral temporal and parietal association cortex bilaterally, and in the right amygdala. Topographically, inflammation co-localized most strongly with tau (r = 0.63 ± 0.24). This correlation was higher than the co-localization of Aβ with tau (r = 0.55 ± 0.25) and of inflammation with Aβ (0.43 ± 0.22). Inflammation co-localized least with atrophy (-0.29 ± 0.26). These regional correlations could be detected in participants with any of the three rs6971 TSPO polymorphisms. Inflammation in Alzheimer's disease-related regions correlated with impaired cognitive scores. Our data highlight the importance of inflammation, a potential therapeutic target, in the Alzheimer's disease process. Furthermore, they support the notion that, as shown in experimental tissue and animal models, the propagation of tau in humans is associated with brain inflammation.
Collapse
Affiliation(s)
- Johanna Appleton
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Quentin Finn
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | | | - Meixiang Yu
- Cyclotron and Radiopharmaceutical Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Alireza Faridar
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Mohammad O Nakawah
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Carlos Zarate
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Maria C Carrillo
- Medical & Scientific Relations Division, Alzheimer's Association, Chicago, IL 60603, USA
| | | | - Gil D Rabinovici
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Cumbers GA, Harvey-Latham ED, Kassiou M, Werry EL, Danon JJ. Emerging TSPO-PET Radiotracers for Imaging Neuroinflammation: A Critical Analysis. Semin Nucl Med 2024; 54:856-874. [PMID: 39477764 DOI: 10.1053/j.semnuclmed.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/19/2024]
Abstract
The translocator protein (TSPO) is a biomarker for imaging neuroinflammation via Positron Emission Tomography (PET) across a broad range of CNS conditions. Most clinically used PET ligands targeting TSPO have limitations, including high lipophilicity and off-target binding or poor binding to a mutated TSPO isoform present in up to 30% of the population. Research efforts over the past decade have focused on development of improved TSPO PET radiotracers that overcome these limitations. This review provides a critical analysis of the development and validation of these so-called "third-generation" radiotracers in clinical and preclinical settings. We also offer our perspective on the future directions of TSPO PET imaging, including recommendations for overcoming current challenges and capitalizing on emerging opportunities in molecular imaging for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Grace A Cumbers
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia
| | - Edward D Harvey-Latham
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia
| | - Michael Kassiou
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia.
| | - Eryn L Werry
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia; Central Clinical School, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Jonathan J Danon
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Salerno S, Viviano M, Baglini E, Poggetti V, Giorgini D, Castagnoli J, Barresi E, Castellano S, Da Settimo F, Taliani S. TSPO Radioligands for Neuroinflammation: An Overview. Molecules 2024; 29:4212. [PMID: 39275061 PMCID: PMC11397380 DOI: 10.3390/molecules29174212] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
The translocator protein (TSPO) is predominately localized on the outer mitochondrial membrane in steroidogenic cells. In the brain, TSPO expression, low under normal conditions, results upregulated in response to glial cell activation, that occurs in neuroinflammation. As a consequence, TSPO has been extensively studied as a biomarker of such conditions by means of TSPO-targeted radiotracers. Although [11C]-PK11195, the prototypical TSPO radioligand, is still widely used for in vivo studies, it is endowed with severe limitations, mainly low sensitivity and poor amenability to quantification. Consequently, several efforts have been focused on the design of new radiotracers for the in vivo imaging of TSPO. The present review will provide an outlook on the latest advances in TSPO radioligands for neuroinflammation imaging. The final goal is to pave the way for (radio)chemists in the future design and development of novel effective and sensitive radiopharmaceuticals targeting TSPO.
Collapse
Affiliation(s)
- Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Monica Viviano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.V.); (D.G.); (S.C.)
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Doralice Giorgini
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.V.); (D.G.); (S.C.)
| | - Jacopo Castagnoli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.V.); (D.G.); (S.C.)
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.S.); (E.B.); (V.P.); (J.C.); (S.T.)
| |
Collapse
|
8
|
Giladi M, Montgomery AP, Kassiou M, Danon JJ. Structure-based drug design for TSPO: Challenges and opportunities. Biochimie 2024; 224:41-50. [PMID: 38782353 DOI: 10.1016/j.biochi.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/27/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
The translocator protein 18 kDa (TSPO) is an evolutionarily conserved mitochondrial transmembrane protein implicated in various neuropathologies and inflammatory conditions, making it a longstanding diagnostic and therapeutic target of interest. Despite the development of various classes of TSPO ligand chemotypes, and the elucidation of bacterial and non-human mammalian experimental structures, many unknowns exist surrounding its differential structural and functional features in health and disease. There are several limitations associated with currently used computational methodologies for modelling the native structure and ligand-binding behaviour of this enigmatic protein. In this perspective, we provide a critical analysis of the developments in the uses of these methods, outlining their uses, inherent limitations, and continuing challenges. We offer suggestions of unexplored opportunities that exist in the use of computational methodologies which offer promise for enhancing our understanding of the TSPO.
Collapse
Affiliation(s)
- Mia Giladi
- School of Chemistry, The University of Sydney, 2050, Sydney, NSW, Australia
| | | | - Michael Kassiou
- School of Chemistry, The University of Sydney, 2050, Sydney, NSW, Australia.
| | - Jonathan J Danon
- School of Chemistry, The University of Sydney, 2050, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Lee N, Choi JY, Ryu YH. The development status of PET radiotracers for evaluating neuroinflammation. Nucl Med Mol Imaging 2024; 58:160-176. [PMID: 38932754 PMCID: PMC11196502 DOI: 10.1007/s13139-023-00831-4] [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: 10/11/2023] [Revised: 11/16/2023] [Accepted: 12/05/2023] [Indexed: 06/28/2024] Open
Abstract
Neuroinflammation is associated with the pathophysiologies of neurodegenerative and psychiatric disorders. Evaluating neuroinflammation using positron emission tomography (PET) plays an important role in the early diagnosis and determination of proper treatment of brain diseases. To quantify neuroinflammatory responses in vivo, many PET tracers have been developed using translocator proteins, imidazole-2 binding site, cyclooxygenase, monoamine oxidase-B, adenosine, cannabinoid, purinergic P2X7, and CSF-1 receptors as biomarkers. In this review, we introduce the latest developments in PET tracers that can image neuroinflammation, focusing on clinical trials, and further consider their current implications.
Collapse
Affiliation(s)
- Namhun Lee
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Bartos LM, Kirchleitner SV, Kolabas ZI, Quach S, Beck A, Lorenz J, Blobner J, Mueller SA, Ulukaya S, Hoeher L, Horvath I, Wind-Mark K, Holzgreve A, Ruf VC, Gold L, Kunze LH, Kunte ST, Beumers P, Park HE, Antons M, Zatcepin A, Briel N, Hoermann L, Schaefer R, Messerer D, Bartenstein P, Riemenschneider MJ, Lindner S, Ziegler S, Herms J, Lichtenthaler SF, Ertürk A, Tonn JC, von Baumgarten L, Albert NL, Brendel M. Deciphering sources of PET signals in the tumor microenvironment of glioblastoma at cellular resolution. SCIENCE ADVANCES 2023; 9:eadi8986. [PMID: 37889970 PMCID: PMC10610915 DOI: 10.1126/sciadv.adi8986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
Various cellular sources hamper interpretation of positron emission tomography (PET) biomarkers in the tumor microenvironment (TME). We developed an approach of immunomagnetic cell sorting after in vivo radiotracer injection (scRadiotracing) with three-dimensional (3D) histology to dissect the cellular allocation of PET signals in the TME. In mice with implanted glioblastoma, translocator protein (TSPO) radiotracer uptake per tumor cell was higher compared to tumor-associated microglia/macrophages (TAMs), validated by protein levels. Translation of in vitro scRadiotracing to patients with glioma immediately after tumor resection confirmed higher single-cell TSPO tracer uptake of tumor cells compared to immune cells. Across species, cellular radiotracer uptake explained the heterogeneity of individual TSPO-PET signals. In consideration of cellular tracer uptake and cell type abundance, tumor cells were the main contributor to TSPO enrichment in glioblastoma; however, proteomics identified potential PET targets highly specific for TAMs. Combining cellular tracer uptake measures with 3D histology facilitates precise allocation of PET signals and serves to validate emerging novel TAM-specific radioligands.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Alexander Beck
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Julia Lorenz
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Jens Blobner
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stephan A. Mueller
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- School of Computation, Information and Technology (CIT), TUM, Boltzmannstr. 3, 85748 Garching, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lukas Gold
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sebastian T. Kunte
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Philipp Beumers
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ha Eun Park
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Melissa Antons
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Nils Briel
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Leonie Hoermann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Rebecca Schaefer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Denise Messerer
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan F. Lichtenthaler
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
11
|
Cools R, Kerkhofs K, Leitao RCF, Bormans G. Preclinical Evaluation of Novel PET Probes for Dementia. Semin Nucl Med 2023; 53:599-629. [PMID: 37149435 DOI: 10.1053/j.semnuclmed.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 05/08/2023]
Abstract
The development of novel PET imaging agents that selectively bind specific dementia-related targets can contribute significantly to accurate, differential and early diagnosis of dementia causing diseases and support the development of therapeutic agents. Consequently, in recent years there has been a growing body of literature describing the development and evaluation of potential new promising PET tracers for dementia. This review article provides a comprehensive overview of novel dementia PET probes under development, classified by their target, and pinpoints their preclinical evaluation pathway, typically involving in silico, in vitro and ex/in vivo evaluation. Specific target-associated challenges and pitfalls, requiring extensive and well-designed preclinical experimental evaluation assays to enable successful clinical translation and avoid shortcomings observed for previously developed 'well-established' dementia PET tracers are highlighted in this review.
Collapse
Affiliation(s)
- Romy Cools
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Kobe Kerkhofs
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; NURA, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Renan C F Leitao
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Cao Y, Yue X, Jia M, Wang J. Neuroinflammation and anti-inflammatory therapy for ischemic stroke. Heliyon 2023; 9:e17986. [PMID: 37519706 PMCID: PMC10372247 DOI: 10.1016/j.heliyon.2023.e17986] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 04/25/2023] [Accepted: 07/04/2023] [Indexed: 08/01/2023] Open
Abstract
Stroke remains one of the most devastating and challenging neurological diseases worldwide. Inflammation, as well as oxidative stress is one of the main contributors to post-stroke injuries, and oxidative stress can further induce inflammation. Moreover, the inflammatory response is closely related to immune modulation in ischemic stroke progression. Hence, major ischemic stroke treatment strategies include targeting inflammatory responses, immune modulation (especially immune cells), and inflammatory response to suppress stroke progression. To date, several drugs have demonstrated clinical efficacy, such as Etanercept and Fingolimod. However, only edaravone dexborneol has successfully passed the phase III clinical trial and been approved by the National Medical Products Administration (NMPA) to treat ischemic stroke in China, which can restore redox balance and regulate inflammatory immune responses, thus providing neuroprotection in ischemic stroke. In this review, we will comprehensively summarize the current advances in the application of inflammatory biomarkers, neuroinflammation and neuro-immunotherapeutic scenarios for ischemic stroke, thus aiming to provide a theoretical basis and new prospects and frontiers for clinical applications.
Collapse
Affiliation(s)
- Yangyue Cao
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xuanye Yue
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Jia
- National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiawei Wang
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Conte M, De Feo MS, Corica F, Gorica J, Sidrak MMA, De Cristofaro F, Filippi L, Ricci M, De Vincentis G, Frantellizzi V. A Systematic Review on Dementia and Translocator Protein (TSPO): When Nuclear Medicine Highlights an Underlying Expression. Biomolecules 2023; 13:biom13040598. [PMID: 37189346 DOI: 10.3390/biom13040598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Translocator protein (TSPO) is a neuroinflammation hallmark. Different TSPO affinity compounds have been produced and over time, the techniques of radiolabeling have been refined. The aim of this systematic review is to summarize the development of new radiotracers for dementia and neuroinflammation imaging. Methods: An online search of the literature was conducted in the PubMed, Scopus, Medline, Cochrane Library, and Web of Science databases, selecting published studies from January 2004 to December 2022. The accepted studies considered the synthesis of TSPO tracers for nuclear medicine imaging in dementia and neuroinflammation. Results: A total of 50 articles was identified. Twelve papers were selected from the included studies’ bibliographies and 34 were excluded. Thus, 28 articles were ultimately selected for quality assessment. Conclusion: Huge efforts in developing specific and stable tracers for PET/SPECT imaging have been made. The long half-life of 18F makes this isotope a preferable choice to 11C. An emerging limitation to this however is that neuroinflammation involves all of the brain which inhibits the possibility of detecting a slight inflammation status change in patients. A partial solution to this is using the cerebellum as a reference region and developing higher TSPO affinity tracers. Moreover, it is necessary to consider the presence of distomers and racemic compounds interfering with pharmacological tracers’ effects and increasing the noise ratio in images.
Collapse
|
14
|
Tan Z, Haider A, Zhang S, Chen J, Wei J, Liao K, Li G, Wei H, Dong C, Ran W, Li Y, Li Y, Rong J, Li Y, Liang SH, Xu H, Wang L. Quantitative assessment of translocator protein (TSPO) in the non-human primate brain and clinical translation of [ 18F]LW223 as a TSPO-targeted PET radioligand. Pharmacol Res 2023; 189:106681. [PMID: 36746361 DOI: 10.1016/j.phrs.2023.106681] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Translocator protein 18 kDa (TSPO) positron emission tomography (PET) can be harnessed for the non-invasive detection of macrophage-driven inflammation. [18F]LW223, a newly reported TSPO PET tracer which was insensitive to rs6971 polymorphism, showed favorable performance characteristics in a recent imaging study involving a rat myocardial infarction model. To enable quantitative neuroimaging with [18F]LW223, we conducted kinetic analysis in the non-human primate (NHP) brain. Further, we sought to assess the utility of [18F]LW223-based TSPO imaging in a first-in-human study. METHODS Radiosynthesis of [18F]LW223 was accomplished on an automated module, whereas molar activities, stability in formulation, lipophilicity and unbound free fraction (fu) of the probe were measured. Brain penetration and target specificity of [18F]LW223 in NHPs were corroborated by PET-MR imaging under baseline and pre-blocking conditions using the validated TSPO inhibitor, (R)-PK11195, at doses ranging from 5 to 10 mg/kg. Kinetic modeling was performed using one-tissue compartment model (1TCM), two-tissue compartment model (2TCM) and Logan graphical analyses, using dynamic PET data acquisition, arterial blood collection and metabolic stability testing. Clinical PET scans were performed in two healthy volunteers (HVs). Regional brain standard uptake value ratio (SUVr) was assessed for different time intervals. RESULTS [18F]LW223 was synthesized in non-decay corrected radiochemical yields (n.d.c. RCYs) of 33.3 ± 6.5% with molar activities ranging from 1.8 ± 0.7 Ci/µmol (n = 11). [18F]LW223 was stable in formulation for up to 4 h and LogD7.4 of 2.31 ± 0.13 (n = 6) and fu of 5.80 ± 1.42% (n = 6) were determined. [18F]LW223 exhibited good brain penetration in NHPs, with a peak SUV value of ca. 1.79 in the whole brain. Pre-treatment with (R)-PK11195 substantially accelerated the washout and attenuated the area under the time-activity curve, indicating in vivo specificity of [18F]LW223 towards TSPO. Kinetic modeling demonstrated that 2TCM was the most suitable model for [18F]LW223-based neuroimaging. Global transfer rate constants (K1) and total volumes of distribution (VT) were found to be 0.10 ± 0.01 mL/cm3/min and 2.30 ± 0.17 mL/cm3, respectively. Dynamic PET data analyses across distinct time windows revealed that the VT values were relatively stable after 60 min post-injection. In a preliminary clinical study with two healthy volunteers, [18F]LW223 exhibited good brain uptake and considerable tracer retention across all analyzed brain regions. Of note, an excellent correlation between SUVr with VT was obtained when assessing the time interval from 20 to 40 min post tracer injection (SUVr(20-40 min), R2 = 0.94, p < 0.0001), suggesting this time window may be suitable to estimate specific binding to TSPO in human brain. CONCLUSION Our findings indicate that [18F]LW223 is suitable for quantitative TSPO-targeted PET imaging in higher species. Employing state-of-the-art kinetic modeling, we found that [18F]LW223 was effective in mapping TSPO throughout the NHP brain, with best model fits obtained from 2TCM and Logan graphical analyses. Overall, our results indicate that [18F]LW223 exhibits favorable tracer performance characteristics in higher species, and this novel imaging tool may hold promise to provide effective neuroinflammation imaging in patients with neurological disease.
Collapse
Affiliation(s)
- Zhiqiang Tan
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Achi Haider
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Junjie Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Kai Liao
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Chenchen Dong
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Wenqing Ran
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ying Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yuefeng Li
- Guangdong Landau Biotechnology Co. Ltd., Guangzhou 510555, China
| | - Jian Rong
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA.
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
15
|
Yang Z, Banks SJ, Ritter AR, Cummings JL, Sreenivasan K, Kinney JW, Caldwell JK, Wong CG, Miller JB, Cordes D. Microglial Imaging in Alzheimer's Disease and Its Relationship to Brain Amyloid: A Human 18F-GE180 PET Study. J Alzheimers Dis 2023; 96:1505-1514. [PMID: 37980664 PMCID: PMC10894577 DOI: 10.3233/jad-230631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Emerging evidence suggests a potential causal role of neuroinflammation in Alzheimer's disease (AD). Using positron emission tomography (PET) to image overexpressed 18 kDA translocator protein (TSPO) by activated microglia has gained increasing interest. The uptake of 18F-GE180 TSPO PET was observed to co-localize with inflammatory markers and have a two-stage association with amyloid PET in mice. Very few studies evaluated the diagnostic power of 18F-GE180 PET in AD population and its interpretation in human remains controversial about whether it is a marker of microglial activation or merely reflects disrupted blood-brain barrier integrity in humans. OBJECTIVE The goal of this study was to study human GE180 from the perspective of the previous animal observations. METHODS With data from twenty-four participants having 18F-GE180 and 18F-AV45 PET scans, we evaluated the group differences of 18F-GE180 uptake between participants with and without cognitive impairment. An association analysis of 18F-GE180 and 18F-AV45 was then conducted to test if the relationship in humans is consistent with the two-stage association in AD mouse model. RESULTS Elevated 18F-GE180 was observed in participants with cognitive impairment compared to those with normal cognition. No regions showed reduced 18F-GE180 uptake. Consistent with mouse model, a two-stage association between 18F-GE180 and 18F-AV45 was observed. CONCLUSIONS 18F-GE180 PET imaging showed promising utility in detecting pathological alterations in a symptomatic AD population. Consistent two-stage association between 18F-GE180 and amyloid PET in human and mouse suggested that 18F-GE180 uptake in human might be considerably influenced by microglial activation.
Collapse
Affiliation(s)
- Zhengshi Yang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| | | | - Aaron R. Ritter
- Hoag’s Pickup Family Neurosciences Institute, Newport Beach, CA, USA
| | - Jeffrey L. Cummings
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Karthik Sreenivasan
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Jefferson W. Kinney
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | | | - Christina G. Wong
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Justin B. Miller
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA
| |
Collapse
|
16
|
Keeling G, Man F. Nuclear Imaging of Inflammation. PROGRESS IN INFLAMMATION RESEARCH 2023:23-90. [DOI: 10.1007/978-3-031-23661-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
18F-Radiolabeled Translocator Protein (TSPO) PET Tracers: Recent Development of TSPO Radioligands and Their Application to PET Study. Pharmaceutics 2022; 14:pharmaceutics14112545. [PMID: 36432736 PMCID: PMC9697781 DOI: 10.3390/pharmaceutics14112545] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a transmembrane protein in the mitochondrial membrane, which has been identified as a peripheral benzodiazepine receptor. TSPO is generally present at high concentrations in steroid-producing cells and plays an important role in steroid synthesis, apoptosis, and cell proliferation. In the central nervous system, TSPO expression is relatively modest under normal physiological circumstances. However, some pathological disorders can lead to changes in TSPO expression. Overexpression of TSPO is associated with several diseases, such as neurodegenerative diseases, neuroinflammation, brain injury, and cancers. TSPO has therefore become an effective biomarker of related diseases. Positron emission tomography (PET), a non-invasive molecular imaging technique used for the clinical diagnosis of numerous diseases, can detect diseases related to TSPO expression. Several radiolabeled TSPO ligands have been developed for PET. In this review, we describe recent advances in the development of TSPO ligands, and 18F-radiolabeled TSPO in particular, as PET tracers. This review covers pharmacokinetic studies, preclinical and clinical trials of 18F-labeled TSPO PET ligands, and the synthesis of TSPO ligands.
Collapse
|
18
|
Viviano M, Barresi E, Siméon FG, Costa B, Taliani S, Da Settimo F, Pike VW, Castellano S. Essential Principles and Recent Progress in the Development of TSPO PET Ligands for Neuroinflammation Imaging. Curr Med Chem 2022; 29:4862-4890. [PMID: 35352645 PMCID: PMC10080361 DOI: 10.2174/0929867329666220329204054] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
The translocator protein 18kDa (TSPO) is expressed in the outer mitochondrial membrane and is implicated in several functions, including cholesterol transport and steroidogenesis. Under normal physiological conditions, TSPO is present in very low concentrations in the human brain but is markedly upregulated in response to brain injury and inflammation. This upregulation is strongly associated with activated microglia. Therefore, TSPO is particularly suited for assessing active gliosis associated with brain lesions following injury or disease. For over three decades, TSPO has been studied as a biomarker. Numerous radioligands for positron emission tomography (PET) that target TSPO have been developed for imaging inflammatory progression in the brain. Although [11C]PK11195, the prototypical first-generation PET radioligand, is still widely used for in vivo studies, mainly now as its single more potent R-enantiomer, it has severe limitations, including low sensitivity and poor amenability to quantification. Second-generation radioligands are characterized by higher TSPO specific signals but suffer from other drawbacks, such as sensitivity to the TSPO single nucleotide polymorphism (SNP) rs6971. Therefore, their applications in human studies have the burden of needing to genotype subjects. Consequently, recent efforts are focused on developing improved radioligands that combine the optimal features of the second generation with the ability to overcome the differences in binding affinities across the population. This review presents essential principles in the design and development of TSPO PET ligands and discusses prominent examples among the main chemotypes.
Collapse
Affiliation(s)
- Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | | | - Fabrice G. Siméon
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | | | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| |
Collapse
|
19
|
Neuroinflammation in Low-Level PM2.5-Exposed Rats Illustrated by PET via an Improved Automated Produced [18F]FEPPA: A Feasibility Study. Mol Imaging 2022; 2022:1076444. [PMID: 35903248 PMCID: PMC9328187 DOI: 10.1155/2022/1076444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/10/2022] [Accepted: 05/18/2022] [Indexed: 11/26/2022] Open
Abstract
Background [18F]FEPPA is a potent TSPO imaging agent that has been found to be a potential tracer for imaging neuroinflammation. In order to fulfill the demand of this tracer for preclinical and clinical studies, we have developed a one-pot automated synthesis with simplified HPLC purification of this tracer, which was then used for PET imaging of neuroinflammation in fine particulate matter- (PM2.5-) exposed rats. Results Using this automated synthesis method, the RCY of the [18F]FEPPA was 38 ± 4% (n = 17, EOB) in a synthesis time of 83 ± 8 min from EOB. The radiochemical purity and molar activities were greater than 99% and 209 ± 138 GBq/μmol (EOS, n = 15), respectively. The quality of the [18F]FEPPA synthesized by this method met the U.S. Pharmacopoeia (USP) criteria. The stability test showed that the [18F]FEPPA was stable at 21 ± 2°C for up to 4 hr after the end of synthesis (EOS). Moreover, microPET imaging showed that increased tracer activity of [18F]FEPPA in the brain of PM2.5-exposed rats (n = 6) were higher than that of normal controls (n = 6) and regional-specific. Conclusions Using the improved semipreparative HPLC purification, [18F]FEPPA has been produced in high quantity, high quality, and high reproducibility and, for the first time, used for PET imaging the effects of PM2.5 in the rat brain. It is ready to be used for imaging inflammation in various clinical or preclinical studies, especially for nearby PET centers without cyclotrons.
Collapse
|
20
|
van der Geest KSM, Sandovici M, Nienhuis PH, Slart RHJA, Heeringa P, Brouwer E, Jiemy WF. Novel PET Imaging of Inflammatory Targets and Cells for the Diagnosis and Monitoring of Giant Cell Arteritis and Polymyalgia Rheumatica. Front Med (Lausanne) 2022; 9:902155. [PMID: 35733858 PMCID: PMC9207253 DOI: 10.3389/fmed.2022.902155] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/13/2022] [Indexed: 12/26/2022] Open
Abstract
Giant cell arteritis (GCA) and polymyalgia rheumatica (PMR) are two interrelated inflammatory diseases affecting patients above 50 years of age. Patients with GCA suffer from granulomatous inflammation of medium- to large-sized arteries. This inflammation can lead to severe ischemic complications (e.g., irreversible vision loss and stroke) and aneurysm-related complications (such as aortic dissection). On the other hand, patients suffering from PMR present with proximal stiffness and pain due to inflammation of the shoulder and pelvic girdles. PMR is observed in 40-60% of patients with GCA, while up to 21% of patients suffering from PMR are also affected by GCA. Due to the risk of ischemic complications, GCA has to be promptly treated upon clinical suspicion. The treatment of both GCA and PMR still heavily relies on glucocorticoids (GCs), although novel targeted therapies are emerging. Imaging has a central position in the diagnosis of GCA and PMR. While [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET) has proven to be a valuable tool for diagnosis of GCA and PMR, it possesses major drawbacks such as unspecific uptake in cells with high glucose metabolism, high background activity in several non-target organs and a decrease of diagnostic accuracy already after a short course of GC treatment. In recent years, our understanding of the immunopathogenesis of GCA and, to some extent, PMR has advanced. In this review, we summarize the current knowledge on the cellular heterogeneity in the immunopathology of GCA/PMR and discuss how recent advances in specific tissue infiltrating leukocyte and stromal cell profiles may be exploited as a source of novel targets for imaging. Finally, we discuss prospective novel PET radiotracers that may be useful for the diagnosis and treatment monitoring in GCA and PMR.
Collapse
Affiliation(s)
- Kornelis S. M. van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Pieter H. Nienhuis
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - William F. Jiemy
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
21
|
Masdeu JC, Pascual B, Fujita M. Imaging Neuroinflammation in Neurodegenerative Disorders. J Nucl Med 2022; 63:45S-52S. [PMID: 35649654 DOI: 10.2967/jnumed.121.263200] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation plays a major role in the etiopathology of neurodegenerative diseases, including Alzheimer and Parkinson diseases, frontotemporal lobar degeneration, and amyotrophic lateral sclerosis. In vivo monitoring of neuroinflammation using PET is critical to understand this process, and data are accumulating in this regard, thus a review is useful. From PubMed, we retrieved publications using any of the available PET tracers to image neuroinflammation in humans as well as selected articles dealing with experimental animal models or the chemistry of currently used or potential radiotracers. We reviewed 280 articles. The most common PET neuroinflammation target, translocator protein (TSPO), has limitations, lacking cellular specificity and the ability to separate neuroprotective from neurotoxic inflammation. However, TSPO PET is useful to define the amount and location of inflammation in the brain of people with neurodegenerative disorders. We describe the characteristics of TSPO and other potential PET neuroinflammation targets and PET tracers available or in development. Despite target and tracer limitations, in recent years there has been a sharp increase in the number of reports of neuroinflammation PET in humans. The most studied has been Alzheimer disease, in which neuroinflammation seems initially neuroprotective and neurotoxic later in the progression of the disease. We describe the findings in all the major neurodegenerative disorders. Neuroinflammation PET is an indispensable tool to understand the process of neurodegeneration, particularly in humans, as well as to validate target engagement in therapeutic clinical trials.
Collapse
Affiliation(s)
- Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and
| | - Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and
| | - Masahiro Fujita
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and.,PET Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas
| |
Collapse
|
22
|
18F-GE180, a failed tracer for translocator protein, has no place in child abuse imaging. Pediatr Radiol 2022; 52:1015-1016. [PMID: 34837109 PMCID: PMC9035024 DOI: 10.1007/s00247-021-05248-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/16/2021] [Accepted: 11/15/2021] [Indexed: 10/19/2022]
|
23
|
Dirks M, Buchert R, Wirries AK, Pflugrad H, Grosse GM, Petrusch C, Schütze C, Wilke F, Mamach M, Hamann L, Langer LBN, Ding XQ, Barg-Hock H, Klempnauer J, Wetzel CH, Lukacevic M, Janssen E, Kessler M, Bengel FM, Geworski L, Rupprecht R, Ross TL, Berding G, Weissenborn K. Reduced microglia activity in patients with long-term immunosuppressive therapy after liver transplantation. Eur J Nucl Med Mol Imaging 2021; 49:234-245. [PMID: 33978829 PMCID: PMC8712291 DOI: 10.1007/s00259-021-05398-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/02/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Calcineurin inhibitors (CNI) can cause long-term impairment of brain function. Possible pathomechanisms include alterations of the cerebral immune system. This study used positron emission tomography (PET) imaging with the translocator protein (TSPO) ligand 18F-GE-180 to evaluate microglial activation in liver-transplanted patients under different regimens of immunosuppression. METHODS PET was performed in 22 liver-transplanted patients (3 CNI free, 9 with low-dose CNI, 10 with standard-dose CNI immunosuppression) and 9 healthy controls. The total distribution volume (VT) estimated in 12 volumes-of-interest was analyzed regarding TSPO genotype, CNI therapy, and cognitive performance. RESULTS In controls, VT was about 80% higher in high affinity binders (n = 5) compared to mixed affinity binders (n = 3). Mean VT corrected for TSPO genotype was significantly lower in patients compared to controls, especially in patients in whom CNI dose had been reduced because of nephrotoxic side effect. CONCLUSION Our results provide evidence of chronic suppression of microglial activity in liver-transplanted patients under CNI therapy especially in patients with high sensitivity to CNI toxicity.
Collapse
Affiliation(s)
- Meike Dirks
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- Integrated Research and Treatment Centre Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany.
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ann-Katrin Wirries
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Henning Pflugrad
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Integrated Research and Treatment Centre Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | - Gerrit M Grosse
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Carlotta Petrusch
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Christian Schütze
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Florian Wilke
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Martin Mamach
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Linda Hamann
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Laura B N Langer
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Xiao-Qi Ding
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Hannelore Barg-Hock
- General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Jürgen Klempnauer
- General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Mario Lukacevic
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Eike Janssen
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Mariella Kessler
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Lilli Geworski
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Georg Berding
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Integrated Research and Treatment Centre Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| |
Collapse
|
24
|
Wimberley C, Lavisse S, Hillmer A, Hinz R, Turkheimer F, Zanotti-Fregonara P. Kinetic modeling and parameter estimation of TSPO PET imaging in the human brain. Eur J Nucl Med Mol Imaging 2021; 49:246-256. [PMID: 33693967 PMCID: PMC8712306 DOI: 10.1007/s00259-021-05248-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/07/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Translocator protein 18-kDa (TSPO) imaging with positron emission tomography (PET) is widely used in research studies of brain diseases that have a neuro-immune component. Quantification of TSPO PET images, however, is associated with several challenges, such as the lack of a reference region, a genetic polymorphism affecting the affinity of the ligand for TSPO, and a strong TSPO signal in the endothelium of the brain vessels. These challenges have created an ongoing debate in the field about which type of quantification is most useful and whether there is an appropriate simplified model. METHODS This review focuses on the quantification of TSPO radioligands in the human brain. The various methods of quantification are summarized, including the gold standard of compartmental modeling with metabolite-corrected input function as well as various alternative models and non-invasive approaches. Their advantages and drawbacks are critically assessed. RESULTS AND CONCLUSIONS Researchers employing quantification methods for TSPO should understand the advantages and limitations associated with each method. Suggestions are given to help researchers choose between these viable alternative methods.
Collapse
Affiliation(s)
| | - Sonia Lavisse
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Departments of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Departments of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, USA
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, M20 3LJ, UK
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Centre for Neuroimaging Sciences, King's College London, De Crespigny Park, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, UK
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Lee SH, Denora N, Laquintana V, Mangiatordi GF, Lopedota A, Lopalco A, Cutrignelli A, Franco M, Delre P, Song IH, Kim HW, Kim SB, Park HS, Kim K, Lee SY, Youn H, Lee BC, Kim SE. Radiosynthesis and characterization of [ 18F]BS224: a next-generation TSPO PET ligand insensitive to the rs6971 polymorphism. Eur J Nucl Med Mol Imaging 2021; 49:110-124. [PMID: 34783879 PMCID: PMC8712300 DOI: 10.1007/s00259-021-05617-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Translocator protein 18-kDa (TSPO) positron emission tomography (PET) is a valuable tool to detect neuroinflammed areas in a broad spectrum of neurodegenerative diseases. However, the clinical application of second-generation TSPO ligands as biomarkers is limited because of the presence of human rs6971 polymorphism that affects their binding. Here, we describe the ability of a new TSPO ligand, [18F]BS224, to identify abnormal TSPO expression in neuroinflammation independent of the rs6971 polymorphism. METHODS An in vitro competitive inhibition assay of BS224 was conducted with [3H]PK 11195 using membrane proteins isolated from 293FT cells expressing TSPO-wild type (WT) or TSPO-mutant A147T (Mut), corresponding to a high-affinity binder (HAB) and low-affinity binder (LAB), respectively. Molecular docking was performed to investigate the interaction of BS224 with the binding sites of rat TSPO-WT and TSPO-Mut. We synthesized a new 18F-labeled imidazopyridine acetamide ([18F]BS224) using boronic acid pinacol ester 6 or iodotoluene tosylate precursor 7, respectively, via aromatic 18F-fluorination. Dynamic PET scanning was performed up to 90 min after the injection of [18F]BS224 to healthy mice, and PET imaging data were obtained to estimate its absorbed doses in organs. To evaluate in vivo TSPO-specific uptake of [18F]BS224, lipopolysaccharide (LPS)-induced inflammatory and ischemic stroke rat models were used. RESULTS BS224 exhibited a high affinity (Ki = 0.51 nM) and selectivity for TSPO. The ratio of IC50 values of BS224 for LAB to that for HAB indicated that the TSPO binding affinity of BS224 has low binding sensitivity to the rs6971 polymorphism and it was comparable to that of PK 11195, which is not sensitive to the polymorphism. Docking simulations showed that the binding mode of BS224 is not affected by the A147T mutation and consequently supported the observed in vitro selectivity of [18F]BS224 regardless of polymorphisms. With optimal radiochemical yield (39 ± 6.8%, decay-corrected) and purity (> 99%), [18F]BS224 provided a clear visible image of the inflammatory lesion with a high signal-to-background ratio in both animal models (BPND = 1.43 ± 0.17 and 1.57 ± 0.37 in the LPS-induced inflammatory and ischemic stroke rat models, respectively) without skull uptake. CONCLUSION Our results suggest that [18F]BS224 may be a promising TSPO ligand to gauge neuroinflammatory disease-related areas in a broad range of patients irrespective of the common rs6971 polymorphism.
Collapse
Affiliation(s)
- Sang Hee Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620 Republic of Korea
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea
| | - Nunzio Denora
- Department of Pharmacy – Drug Sciences, University of Bari “A. Moro”, 70121 Bari, Italy
| | - Valentino Laquintana
- Department of Pharmacy – Drug Sciences, University of Bari “A. Moro”, 70121 Bari, Italy
| | | | - Angela Lopedota
- Department of Pharmacy – Drug Sciences, University of Bari “A. Moro”, 70121 Bari, Italy
| | - Antonio Lopalco
- Department of Pharmacy – Drug Sciences, University of Bari “A. Moro”, 70121 Bari, Italy
| | - Annalisa Cutrignelli
- Department of Pharmacy – Drug Sciences, University of Bari “A. Moro”, 70121 Bari, Italy
| | - Massimo Franco
- Department of Pharmacy – Drug Sciences, University of Bari “A. Moro”, 70121 Bari, Italy
| | - Pietro Delre
- Institute of Crystallography, National Research Council, Via G. Amendola 122/O, 70126 Bari, Italy
- Department of Chemistry, University of Bari “A. Moro”, Via E. Orabona, 4, 70125 Bari, Italy
| | - In Ho Song
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620 Republic of Korea
| | - Hye Won Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620 Republic of Korea
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea
| | - Su Bin Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620 Republic of Korea
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea
| | - Hyun Soo Park
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620 Republic of Korea
| | - Kyungmin Kim
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, 03080 Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080 Republic of Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Seok-Yong Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, 03080 Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080 Republic of Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, 03080 Republic of Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Byung Chul Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620 Republic of Korea
- Center for Nanomolecular Imaging and Innovative Drug Development, Advanced Institutes of Convergence Technology, Suwon, 16229 Republic of Korea
| | - Sang Eun Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620 Republic of Korea
- Center for Nanomolecular Imaging and Innovative Drug Development, Advanced Institutes of Convergence Technology, Suwon, 16229 Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea
| |
Collapse
|
26
|
Sander CY, Bovo S, Torrado-Carvajal A, Albrecht D, Deng H, Napadow V, Price JC, Hooker JM, Loggia ML. [ 11C]PBR28 radiotracer kinetics are not driven by alterations in cerebral blood flow. J Cereb Blood Flow Metab 2021; 41:3069-3084. [PMID: 34159823 PMCID: PMC8756484 DOI: 10.1177/0271678x211023387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The positron emission tomography (PET) radiotracer [11C]PBR28 has been increasingly used to image the translocator protein (TSPO) as a marker of neuroinflammation in a variety of brain disorders. Interrelatedly, similar clinical populations can also exhibit altered brain perfusion, as has been shown using arterial spin labelling in magnetic resonance imaging (MRI) studies. Hence, an unsolved debate has revolved around whether changes in perfusion could alter delivery, uptake, or washout of the radiotracer [11C]PBR28, and thereby influence outcome measures that affect interpretation of TSPO upregulation. In this simultaneous PET/MRI study, we demonstrate that [11C]PBR28 signal elevations in chronic low back pain patients are not accompanied, in the same regions, by increases in cerebral blood flow (CBF) compared to healthy controls, and that areas of marginal hypoperfusion are not accompanied by decreases in [11C]PBR28 signal. In non-human primates, we show that hypercapnia-induced increases in CBF during radiotracer delivery or washout do not alter [11C]PBR28 outcome measures. The combined results from two methodologically distinct experiments provide support from human data and direct experimental evidence from non-human primates that changes in CBF do not influence outcome measures reported by [11C]PBR28 PET imaging studies and corresponding interpretations of the biological meaning of TSPO upregulation.
Collapse
Affiliation(s)
- Christin Y Sander
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Stefano Bovo
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Information Engineering, University of Padova, Padova, Italy
| | - Angel Torrado-Carvajal
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Madrid, Spain
| | - Daniel Albrecht
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Hongping Deng
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Vitaly Napadow
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Julie C Price
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Jacob M Hooker
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Aceves-Serrano L, Sossi V, Doudet DJ. Comparison of Invasive and Non-invasive Estimation of [ 11C]PBR28 Binding in Non-human Primates. Mol Imaging Biol 2021; 24:404-415. [PMID: 34622422 DOI: 10.1007/s11307-021-01661-6] [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: 03/15/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE To identify a reliable alternative to the full blood [11C]PBR28 quantification method that would be easily replicated in multiple research and clinical settings. PROCEDURES Ten [11C]PBR28 scans were acquired from 7 healthy non-human primates (NHP). Arterial input functions (AIFs) were averaged to create a population template input function (TIF). Population-based input functions were created by scaling the TIF with injected activity per body weight (PBIF) or unmetabolized tracer activity in blood at 15-,30-, and 60-min post-injection (PBIF15, PBIF30, and PBIF60). Two additional input functions were used: the native unmetabolized total plasma activity (Totals) and the Totals curve metabolite corrected by a scaled template parent fraction from a 30-min sample (TPF30-IF). Total distribution volumes (VTs) were calculated using PBIF, PBIF30, PBIF15, PBIF60, Totals, TPF30-IF, and the individual AIF (VTAIF). Distribution volume ratios (DVR) were computed using the cerebellum and the centrum semiovale (CSO), as pseudo-reference regions (DVRCereb, DVRCSO). Results obtained with each method were compared to VTAIF. Applicability of these alternative methods was tested on an independent pharmacological challenge dataset of microglial activation and depletion. Evaluation was carried at baseline, immediately after intervention (acute), and weeks post-intervention (post-recovery). RESULTS VTs computed using PBIF15 and PBIF30 showed the best correlation to VTAIF (r > 0.90), while VT derived from the blood-free-scaled PBIF showed poor correlation (r = 0.46) and DVRCSO correlated the least (r = 0.26). In the pharmacological challenge study, most population-derived VT values were comparable to VTAIF at baseline and showed varied sensitivity to challenges at acute and post-recovery evaluation. DVR values did not detect relevant changes. CONCLUSIONS Population-based input functions scaled with a single blood sample might be a useful alternative to using AIF to compute [11C]PBR28 binding in healthy NHPs or animals with comparable metabolism and overall perform better than pseudo-reference regions approaches.
Collapse
Affiliation(s)
- Lucero Aceves-Serrano
- Department of Medicine, Division of Neurology, University of British Columbia, Rm M36 Purdy Pavilion, 2221 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Doris J Doudet
- Department of Medicine, Division of Neurology, University of British Columbia, Rm M36 Purdy Pavilion, 2221 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| |
Collapse
|
28
|
Zhou R, Ji B, Kong Y, Qin L, Ren W, Guan Y, Ni R. PET Imaging of Neuroinflammation in Alzheimer's Disease. Front Immunol 2021; 12:739130. [PMID: 34603323 PMCID: PMC8481830 DOI: 10.3389/fimmu.2021.739130] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation play an important role in Alzheimer's disease pathogenesis. Advances in molecular imaging using positron emission tomography have provided insights into the time course of neuroinflammation and its relation with Alzheimer's disease central pathologies in patients and in animal disease models. Recent single-cell sequencing and transcriptomics indicate dynamic disease-associated microglia and astrocyte profiles in Alzheimer's disease. Mitochondrial 18-kDa translocator protein is the most widely investigated target for neuroinflammation imaging. New generation of translocator protein tracers with improved performance have been developed and evaluated along with tau and amyloid imaging for assessing the disease progression in Alzheimer's disease continuum. Given that translocator protein is not exclusively expressed in glia, alternative targets are under rapid development, such as monoamine oxidase B, matrix metalloproteinases, colony-stimulating factor 1 receptor, imidazoline-2 binding sites, cyclooxygenase, cannabinoid-2 receptor, purinergic P2X7 receptor, P2Y12 receptor, the fractalkine receptor, triggering receptor expressed on myeloid cells 2, and receptor for advanced glycation end products. Promising targets should demonstrate a higher specificity for cellular locations with exclusive expression in microglia or astrocyte and activation status (pro- or anti-inflammatory) with highly specific ligand to enable in vivo brain imaging. In this review, we summarised recent advances in the development of neuroinflammation imaging tracers and provided an outlook for promising targets in the future.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Kong
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Limei Qin
- Inner Mongolia Baicaotang Qin Chinese Mongolia Hospital, Hohhot, China
| | - Wuwei Ren
- School of Information Science and Technology, Shanghaitech University, Shanghai, China
| | - Yihui Guan
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & Eidgenössische Technische Hochschule Zürich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
29
|
De Marchi F, Munitic I, Amedei A, Berry JD, Feldman EL, Aronica E, Nardo G, Van Weehaeghe D, Niccolai E, Prtenjaca N, Sakowski SA, Bendotti C, Mazzini L. Interplay between immunity and amyotrophic lateral sclerosis: Clinical impact. Neurosci Biobehav Rev 2021; 127:958-978. [PMID: 34153344 PMCID: PMC8428677 DOI: 10.1016/j.neubiorev.2021.06.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/07/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating and rapidly fatal neurodegenerative disease. Despite decades of research and many new insights into disease biology over the 150 years since the disease was first described, causative pathogenic mechanisms in ALS remain poorly understood, especially in sporadic cases. Our understanding of the role of the immune system in ALS pathophysiology, however, is rapidly expanding. The aim of this manuscript is to summarize the recent advances regarding the immune system involvement in ALS, with particular attention to clinical translation. We focus on the potential pathophysiologic mechanism of the immune system in ALS, discussing local and systemic factors (blood, cerebrospinal fluid, and microbiota) that influence ALS onset and progression in animal models and people. We also explore the potential of Positron Emission Tomography to detect neuroinflammation in vivo, and discuss ongoing clinical trials of therapies targeting the immune system. With validation in human patients, new evidence in this emerging field will serve to identify novel therapeutic targets and provide realistic hope for personalized treatment strategies.
Collapse
Affiliation(s)
- Fabiola De Marchi
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, Novara, 28100, Italy
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000, Rijeka, Croatia
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - James D Berry
- Sean M. Healey & AMG Center for ALS, Department of Neurology, Massachusetts General Hospital, 165 Cambridge Street, Suite 600, Boston, MA, 02114, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milanm, 20156, Italy
| | - Donatienne Van Weehaeghe
- Division of Nuclear Medicine, Department of Imaging and Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Nikolina Prtenjaca
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000, Rijeka, Croatia
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milanm, 20156, Italy
| | - Letizia Mazzini
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, Novara, 28100, Italy.
| |
Collapse
|
30
|
Giordani A, Menziani MC, Moresco RM, Matarrese M, Paolino M, Saletti M, Giuliani G, Anzini M, Cappelli A. Exploring Translocator Protein (TSPO) Medicinal Chemistry: An Approach for Targeting Radionuclides and Boron Atoms to Mitochondria. J Med Chem 2021; 64:9649-9676. [PMID: 34254805 DOI: 10.1021/acs.jmedchem.1c00379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Translocator protein 18 kDa [TSPO or peripheral-type benzodiazepine receptor (PBR)] was identified in the search of binding sites for benzodiazepine anxiolytic drugs in peripheral regions. In these areas, binding sites for TSPO ligands were recognized in steroid-producing tissues. TSPO plays an important role in many cellular functions, and its coding sequence is highly conserved across species. TSPO is located predominantly on the membrane of mitochondria and is overexpressed in several solid cancers. TSPO basal expression in the CNS is low, but it becomes high in neurodegenerative conditions. Thus, TSPO constitutes not only as an outstanding drug target but also as a valuable marker for the diagnosis of a number of diseases. The aim of the present article is to show the lesson we have learned from our activity in TSPO medicinal chemistry and in approaching the targeted delivery to mitochondria by means of TSPO ligands.
Collapse
Affiliation(s)
- Antonio Giordani
- Rottapharm Biotech S.p.A., Via Valosa di Sopra 9, 20900 Monza, Italy
| | - Maria Cristina Menziani
- Dipartimento di Scienze Chimiche e Geologiche, Università di Modena e Reggio Emilia, Via Campi 103, 41121 Modena, Italy
| | - Rosa Maria Moresco
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Mario Matarrese
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Germano Giuliani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Maurizio Anzini
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
31
|
Vettermann FJ, Harris S, Schmitt J, Unterrainer M, Lindner S, Rauchmann BS, Palleis C, Weidinger E, Beyer L, Eckenweber F, Schuster S, Biechele G, Ferschmann C, Milenkovic VM, Wetzel CH, Rupprecht R, Janowitz D, Buerger K, Perneczky R, Höglinger GU, Levin J, Haass C, Tonn JC, Niyazi M, Bartenstein P, Albert NL, Brendel M. Impact of TSPO Receptor Polymorphism on [ 18F]GE-180 Binding in Healthy Brain and Pseudo-Reference Regions of Neurooncological and Neurodegenerative Disorders. Life (Basel) 2021; 11:484. [PMID: 34073557 PMCID: PMC8229996 DOI: 10.3390/life11060484] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022] Open
Abstract
TSPO-PET tracers are sensitive to a single-nucleotide polymorphism (rs6971-SNP), resulting in low-, medium- and high-affinity binders (LABs, MABs and HABS), but the clinical relevance of [18F]GE-180 is still unclear. We evaluated the impact of rs6971-SNP on in vivo [18F]GE-180 binding in a healthy brain and in pseudo-reference tissue in neuro-oncological and neurodegenerative diseases. Standardized uptake values (SUVs) of [18F]GE-180-PET were assessed using a manually drawn region of interest in the frontoparietal and cerebellar hemispheres. The SUVs were compared between the LABs, MABs and HABs in control, glioma, four-repeat tauopathy (4RT) and Alzheimer's disease (AD) subjects. Second, the SUVs were compared between the patients and controls within their rs6971-subgroups. After excluding patients with prior therapy, 24 LABs (7 control, 5 glioma, 6 4RT and 6 AD) were analyzed. Age- and sex-matched MABs (n = 38) and HABs (n = 50) were selected. The LABs had lower frontoparietal and cerebellar SUVs when compared with the MABs and HABs, but no significant difference was observed between the MABs and HABs. Within each rs6971 group, no SUV difference between the patients and controls was detected in the pseudo-reference tissues. The rs6971-SNP affects [18F]GE-180 quantification, revealing lower binding in the LABs when compared to the MABs and HABs. The frontoparietal and cerebellar ROIs were successfully validated as pseudo-reference regions.
Collapse
Affiliation(s)
- Franziska J Vettermann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Julia Schmitt
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Radiology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Endy Weidinger
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Christian Ferschmann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College, London SW7 2AZ, UK
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg, Germany
| | - Joerg C Tonn
- Department of Neurosurgery, University Hospital of Munich, 81377 Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| |
Collapse
|
32
|
Tolvanen T, Kalliokoski K, Malaspina S, Kuisma A, Lahdenpohja S, Postema EJ, Miller MP, Scheinin M. Safety, Biodistribution, and Radiation Dosimetry of 18F-rhPSMA-7.3 in Healthy Adult Volunteers. J Nucl Med 2021; 62:679-684. [PMID: 33067338 PMCID: PMC8844263 DOI: 10.2967/jnumed.120.252114] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
This first-in-humans study investigated the safety, biodistribution, and radiation dosimetry of a novel 18F-labeled radiohybrid prostate-specific membrane antigen (rhPSMA) PET imaging agent, 18F-rhPSMA-7.3. Methods: Six healthy volunteers (3 men, 3 women) underwent multiple whole-body PET acquisitions at scheduled time points up to 248 min after the administration of 18F-rhPSMA-7.3 (mean activity, 220; range, 210–228 MBq). PET scans were conducted in 3 separate sessions, and subjects were encouraged to void between sessions. Blood and urine samples were collected for up to 4 h after injection to assess metabolite-corrected radioactivity in whole blood, plasma, and urine. Quantitative measurements of 18F radioactivity in volumes of interest over target organs were determined directly from the PET images at 8 time points, and normalized time–activity concentration curves were generated. These normalized cumulated activities were then inputted into the OLINDA/EXM package to calculate the internal radiation dosimetry and the subjects’ effective dose. Results:18F-rhPSMA-7.3 was well tolerated. One adverse event (mild headache, not requiring medication) was considered possibly related to 18F-rhPSMA-7.3. The calculated effective dose was 0.0141 mSv/MBq when using a 3.5-h voiding interval. The organs with the highest mean absorbed dose per unit of administered radioactivity were the adrenals (0.1835 mSv/MBq), the kidneys (0.1722 mSv/MBq), the submandibular glands (0.1479 mSv), and the parotid glands (0.1137 mSv/MBq). At the end of the first scanning session (mean time, 111 min after injection), an average of 7.2% (range, 4.4%–9.0%) of the injected radioactivity of 18F-rhPSMA-7.3 was excreted into urine. Conclusion: The safety, biodistribution, and internal radiation dosimetry of 18F-rhPSMA-7.3 are considered favorable for PET imaging.
Collapse
Affiliation(s)
- Tuula Tolvanen
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Kari Kalliokoski
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Simona Malaspina
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Anna Kuisma
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland.,Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | - Salla Lahdenpohja
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | | | | | - Mika Scheinin
- Clinical Research Services Turku-CRST Ltd., Turku, Finland
| |
Collapse
|
33
|
MacAskill MG, Stadulyte A, Williams L, Morgan TEF, Sloan NL, Alcaide-Corral CJ, Walton T, Wimberley C, McKenzie CA, Spath N, Mungall W, BouHaidar R, Dweck MR, Gray GA, Newby DE, Lucatelli C, Sutherland A, Pimlott SL, Tavares AAS. Quantification of Macrophage-Driven Inflammation During Myocardial Infarction with 18F-LW223, a Novel TSPO Radiotracer with Binding Independent of the rs6971 Human Polymorphism. J Nucl Med 2021; 62:536-544. [PMID: 32859708 PMCID: PMC8049364 DOI: 10.2967/jnumed.120.243600] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/28/2020] [Indexed: 01/09/2023] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of death worldwide, and inflammation is central to tissue response and patient outcomes. The 18-kDa translocator protein (TSPO) has been used in PET as an inflammatory biomarker. The aims of this study were to screen novel, fluorinated, TSPO radiotracers for susceptibility to the rs6971 genetic polymorphism using in vitro competition binding assays in human brain and heart; assess whether the in vivo characteristics of our lead radiotracer, 18F-LW223, are suitable for clinical translation; and validate whether 18F-LW223 can detect macrophage-driven inflammation in a rat MI model. Methods: Fifty-one human brain and 29 human heart tissue samples were screened for the rs6971 polymorphism. Competition binding assays were conducted with 3H-PK11195 and the following ligands: PK11195, PBR28, and our novel compounds (AB5186 and LW223). Naïve rats and mice were used for in vivo PET kinetic studies, radiometabolite studies, and dosimetry experiments. Rats underwent permanent coronary artery ligation and were scanned using PET/CT with an invasive input function at 7 d after MI. For quantification of PET signal in the hypoperfused myocardium, K1 (rate constant for transfer from arterial plasma to tissues) was used as a surrogate marker of perfusion to correct the binding potential for impaired radiotracer transfer from plasma to tissue (BPTC). Results: LW223 binding to TSPO was not susceptible to the rs6971 genetic polymorphism in human brain and heart samples. In rodents, 18F-LW223 displayed a specific uptake consistent with TSPO expression, a slow metabolism in blood (69% of parent at 120 min), a high plasma free fraction of 38.5%, and a suitable dosimetry profile (effective dose of 20.5-24.5 μSv/MBq). 18F-LW223 BPTC was significantly higher in the MI cohort within the infarct territory of the anterior wall relative to the anterior wall of naïve animals (32.7 ± 5.0 vs. 10.0 ± 2.4 cm3/mL/min, P ≤ 0.001). Ex vivo immunofluorescent staining for TSPO and CD68 (macrophage marker) resulted in the same pattern seen with in vivo BPTC analysis. Conclusion:18F-LW223 is not susceptible to the rs6971 genetic polymorphism in in vitro assays, has favorable in vivo characteristics, and is able to accurately map macrophage-driven inflammation after MI.
Collapse
Affiliation(s)
- Mark G MacAskill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | - Agne Stadulyte
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | - Lewis Williams
- School of Chemistry, WestCHEM, University of Glasgow, Glasgow, United Kingdom
| | - Timaeus E F Morgan
- School of Chemistry, WestCHEM, University of Glasgow, Glasgow, United Kingdom
| | - Nikki L Sloan
- School of Chemistry, WestCHEM, University of Glasgow, Glasgow, United Kingdom
| | - Carlos J Alcaide-Corral
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | - Tashfeen Walton
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | - Catriona Wimberley
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Chris-Anne McKenzie
- MRC Edinburgh Brain Tissue Bank, University of Edinburgh, Edinburgh, United Kingdom
| | - Nick Spath
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - William Mungall
- Bioresearch and Veterinary Services, University of Edinburgh, Edinburgh, United Kingdom
| | - Ralph BouHaidar
- Forensic Pathology, University of Edinburgh, Edinburgh, United Kingdom
| | - Marc R Dweck
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A Gray
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Andrew Sutherland
- School of Chemistry, WestCHEM, University of Glasgow, Glasgow, United Kingdom
| | - Sally L Pimlott
- School of Medicine, University of Glasgow, Glasgow, United Kingdom; and
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Adriana A S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Mahler C, Schumacher AM, Unterrainer M, Kaiser L, Höllbacher T, Lindner S, Havla J, Ertl-Wagner B, Patzig M, Seelos K, Neitzel J, Mäurer M, Krumbholz M, Metz I, Brück W, Stadelmann C, Merkler D, Gass A, Milenkovic V, Bartenstein P, Albert NL, Kümpfel T, Kerschensteiner M. TSPO PET imaging of natalizumab-associated progressive multifocal leukoencephalopathy. Brain 2021; 144:2683-2695. [PMID: 33757118 DOI: 10.1093/brain/awab127] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 02/10/2021] [Accepted: 03/01/2021] [Indexed: 01/31/2023] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a severe infection of the central nervous system caused by the polyomavirus JC (JCV) that can occur in multiple sclerosis (MS) patients treated with natalizumab. Clinical management of patients with natalizumab-associated PML is challenging not the least because current imaging tools for the early detection, longitudinal monitoring and differential diagnosis of PML lesions are limited. Here we evaluate whether TSPO positron emission tomography (PET) imaging can be applied to monitor the inflammatory activity of PML lesions over time and differentiate them from MS lesions. For this monocenter pilot study we followed 8 patients with natalizumab-associated PML with PET imaging using the TSPO radioligand [18F]GE-180 combined with frequent 3 T MRI imaging. In addition we compared TSPO PET signals in PML lesions with the signal pattern of MS lesions from 17 independent MS patients. We evaluated the standardized uptake value ratio (SUVR) as well as the morphometry of the TSPO uptake for putative PML and MS lesions areas compared to a radiologically unaffected pseudo-reference region in the cerebrum. Furthermore TSPO expression in situ was immunohistochemically verified by determining the density and cellular identity of TSPO-expressing cells in brain sections from four patients with early natalizumab-associated PML as well as five patients with other forms of PML and six patients with inflammatory demyelinating CNS lesions (clinically isolated syndrome/MS). Histological analysis revealed a reticular accumulation of TSPO expressing phagocytes in PML lesions, while such phagocytes showed a more homogenous distribution in putative MS lesions. TSPO PET imaging showed an enhanced tracer uptake in natalizumab-associated PML lesions that was present from the early to the chronic stages (up to 52 months after PML diagnosis). While gadolinium enhancement on MRI rapidly declined to baseline levels, TSPO tracer uptake followed a slow one phase decay curve. A TSPO-based 3-dimensional diagnostic matrix taking into account the uptake levels as well as the shape and texture of the TSPO signal differentiated more than 96% of PML and MS lesions. Indeed, treatment with rituximab after natalizumab-associated PML in three patients did not affect tracer uptake in the assigned PML lesions but reverted tracer uptake to baseline in the assigned active MS lesions. Taken together our study suggests that TSPO PET imaging can reveal CNS inflammation in natalizumab-associated PML. TSPO PET may facilitate longitudinal monitoring of disease activity and help to distinguish recurrent MS activity from PML progression.
Collapse
Affiliation(s)
- Christoph Mahler
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany.,Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried, Germany
| | - Adrian-Minh Schumacher
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany.,Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Thomas Höllbacher
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany.,Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried, Germany
| | - Birgit Ertl-Wagner
- Institute of Clinical Radiology, University Hospital Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Maximilian Patzig
- Institute of Neuroradiology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Klaus Seelos
- Institute of Neuroradiology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Julia Neitzel
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | | | - Markus Krumbholz
- Department of Neurology & Stroke and Hertie-Institute for Clinical Brain Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Imke Metz
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Doron Merkler
- Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland.,Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Achim Gass
- Department of Neurology, University Hospital Mannheim, Mannheim, Germany
| | - Vladimir Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany.,Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany.,Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
35
|
Rivière G, Jaipuria G, Andreas LB, Leonov A, Giller K, Becker S, Zweckstetter M. Membrane-embedded TSPO: an NMR view. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2021; 50:173-180. [PMID: 33354729 PMCID: PMC8071791 DOI: 10.1007/s00249-020-01487-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/19/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022]
Abstract
Translocator Protein (18 kDa) (TSPO) is a mitochondrial transmembrane protein commonly used as a biomarker for neuroinflammation and is also a potential therapeutic target in neurodegenerative diseases. Despite intensive research efforts, the function of TSPO is still largely enigmatic. Deciphering TSPO structure in the native lipid environment is essential to gain insight into its cellular activities and to design improved diagnostic and therapeutic ligands. Here, we discuss the influence of lipid composition on the structure of mammalian TSPO embedded into lipid bilayers on the basis of solid-state NMR experiments. We further highlight that cholesterol can influence both the tertiary and quaternary TSPO structure and also influence TSPO localization in mitochondria-associated endoplasmic reticulum membranes.
Collapse
Affiliation(s)
- Gwladys Rivière
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Garima Jaipuria
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Loren B Andreas
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Andrei Leonov
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Karin Giller
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Stefan Becker
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Markus Zweckstetter
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany.
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
36
|
Zhang L, Hu K, Shao T, Hou L, Zhang S, Ye W, Josephson L, Meyer JH, Zhang MR, Vasdev N, Wang J, Xu H, Wang L, Liang SH. Recent developments on PET radiotracers for TSPO and their applications in neuroimaging. Acta Pharm Sin B 2021; 11:373-393. [PMID: 33643818 PMCID: PMC7893127 DOI: 10.1016/j.apsb.2020.08.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/15/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is predominately localized to the outer mitochondrial membrane in steroidogenic cells. Brain TSPO expression is relatively low under physiological conditions, but is upregulated in response to glial cell activation. As the primary index of neuroinflammation, TSPO is implicated in the pathogenesis and progression of numerous neuropsychiatric disorders and neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), major depressive disorder (MDD) and obsessive compulsive disorder (OCD). In this context, numerous TSPO-targeted positron emission tomography (PET) tracers have been developed. Among them, several radioligands have advanced to clinical research studies. In this review, we will overview the recent development of TSPO PET tracers, focusing on the radioligand design, radioisotope labeling, pharmacokinetics, and PET imaging evaluation. Additionally, we will consider current limitations, as well as translational potential for future application of TSPO radiopharmaceuticals. This review aims to not only present the challenges in current TSPO PET imaging, but to also provide a new perspective on TSPO targeted PET tracer discovery efforts. Addressing these challenges will facilitate the translation of TSPO in clinical studies of neuroinflammation associated with central nervous system diseases.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
- ANT, adenine nucleotide transporter
- Am, molar activities
- BBB, blood‒brain barrier
- BMSC, bone marrow stromal cells
- BP, binding potential
- BPND, non-displaceable binding potential
- BcTSPO, Bacillus cereus TSPO
- CBD, corticobasal degeneration
- CNS disorders
- CNS, central nervous system
- CRAC, cholesterol recognition amino acid consensus sequence
- DLB, Lewy body dementias
- EP, epilepsy
- FTD, frontotemporal dementia
- HAB, high-affinity binding
- HD, Huntington's disease
- HSE, herpes simplex encephalitis
- IMM, inner mitochondrial membrane
- KA, kainic acid
- LAB, low-affinity binding
- LPS, lipopolysaccharide
- MAB, mixed-affinity binding
- MAO-B, monoamine oxidase B
- MCI, mild cognitive impairment
- MDD, major depressive disorder
- MMSE, mini-mental state examination
- MRI, magnetic resonance imaging
- MS, multiple sclerosis
- MSA, multiple system atrophy
- Microglial activation
- NAA/Cr, N-acetylaspartate/creatine
- Neuroinflammation
- OCD, obsessive compulsive disorder
- OMM, outer mitochondrial membrane
- P2X7R, purinergic receptor P2X7
- PAP7, RIa-associated protein
- PBR, peripheral benzodiazepine receptor
- PCA, posterior cortical atrophy
- PD, Parkinson's disease
- PDD, PD dementia
- PET, positron emission tomography
- PKA, protein kinase A
- PRAX-1, PBR-associated protein 1
- PSP, progressive supranuclear palsy
- Positron emission tomography (PET)
- PpIX, protoporphyrin IX
- QA, quinolinic acid
- RCYs, radiochemical yields
- ROS, reactive oxygen species
- RRMS, relapsing remitting multiple sclerosis
- SA, specific activity
- SAH, subarachnoid hemorrhage
- SAR, structure–activity relationship
- SCIDY, spirocyclic iodonium ylide
- SNL, selective neuronal loss
- SNR, signal to noise ratio
- SUV, standard uptake volume
- SUVR, standard uptake volume ratio
- TBAH, tetrabutyl ammonium hydroxide
- TBI, traumatic brain injury
- TLE, temporal lobe epilepsy
- TSPO
- TSPO, translocator protein
- VDAC, voltage-dependent anion channel
- VT, distribution volume
- d.c. RCYs, decay-corrected radiochemical yields
- dMCAO, distal middle cerebral artery occlusion
- fP, plasma free fraction
- n.d.c. RCYs, non-decay-corrected radiochemical yields
- p.i., post-injection
Collapse
Affiliation(s)
- Lingling Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Department of Neurology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Kuan Hu
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Tuo Shao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Weijian Ye
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey H. Meyer
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto ON M5T 1R8, Canada
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto ON M5T 1R8, Canada
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
37
|
Abstract
This article presents an overview of imaging agents for PET that have been applied for research and diagnostic purposes in patients affected by dementia. Classified by the target which the agents visualize, seven groups of tracers can be distinguished, namely radiopharmaceuticals for: (1) Misfolded proteins (ß-amyloid, tau, α-synuclein), (2) Neuroinflammation (overexpression of translocator protein), (3) Elements of the cholinergic system, (4) Elements of monoamine neurotransmitter systems, (5) Synaptic density, (6) Cerebral energy metabolism (glucose transport/ hexokinase), and (7) Various other proteins. This last category contains proteins involved in mechanisms underlying neuroinflammation or cognitive impairment, which may also be potential therapeutic targets. Many receptors belong to this category: AMPA, cannabinoid, colony stimulating factor 1, metabotropic glutamate receptor 1 and 5 (mGluR1, mGluR5), opioid (kappa, mu), purinergic (P2X7, P2Y12), sigma-1, sigma-2, receptor for advanced glycation endproducts, and triggering receptor expressed on myeloid cells-1, besides several enzymes: cyclooxygenase-1 and 2 (COX-1, COX-2), phosphodiesterase-5 and 10 (PDE5, PDE10), and tropomyosin receptor kinase. Significant advances in neuroimaging have been made in the last 15 years. The use of 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) for quantification of regional cerebral glucose metabolism is well-established. Three tracers for ß-amyloid plaques have been approved by the Food and Drug Administration and European Medicines Agency. Several tracers for tau neurofibrillary tangles are already applied in clinical research. Since many novel agents are in the preclinical or experimental stage of development, further advances in nuclear medicine imaging can be expected in the near future. PET studies with established tracers and tracers for novel targets may result in early diagnosis and better classification of neurodegenerative disorders and in accurate monitoring of therapy trials which involve these targets. PET data have prognostic value and may be used to assess the response of the human brain to interventions, or to select the appropriate treatment strategy for an individual patient.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sofia Marcolini
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands
| | - Peter Paul de Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands; University of Antwerp, Born-Bunge Institute, Neurochemistry and Behavior, Campus Drie Eiken, Wilrijk, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands; Ghent University, Ghent, Belgium
| |
Collapse
|
38
|
Ghosh KK, Padmanabhan P, Yang CT, Wang Z, Palanivel M, Ng KC, Lu J, Carlstedt-Duke J, Halldin C, Gulyás B. An In Vivo Study of a Rat Fluid-Percussion-Induced Traumatic Brain Injury Model with [ 11C]PBR28 and [ 18F]flumazenil PET Imaging. Int J Mol Sci 2021; 22:ijms22020951. [PMID: 33477960 PMCID: PMC7835883 DOI: 10.3390/ijms22020951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) modelled by lateral fluid percussion-induction (LFPI) in rats is a widely used experimental rodent model to explore and understand the underlying cellular and molecular alterations in the brain caused by TBI in humans. Current improvements in imaging with positron emission tomography (PET) have made it possible to map certain features of TBI-induced cellular and molecular changes equally in humans and animals. The PET imaging technique is an apt supplement to nanotheranostic-based treatment alternatives that are emerging to tackle TBI. The present study aims to investigate whether the two radioligands, [11C]PBR28 and [18F]flumazenil, are able to accurately quantify in vivo molecular-cellular changes in a rodent TBI-model for two different biochemical targets of the processes. In addition, it serves to observe any palpable variations associated with primary and secondary injury sites, and in the affected versus the contralateral hemispheres. As [11C]PBR28 is a radioligand of the 18 kD translocator protein, the up-regulation of which is coupled to the level of neuroinflammation in the brain, and [18F]flumazenil is a radioligand for GABAA-benzodiazepine receptors, whose level mirrors interneuronal activity and eventually cell death, the use of the two radioligands may reveal two critical features of TBI. An up-regulation in the [11C]PBR28 uptake triggered by the LFP in the injured (right) hemisphere was noted on day 14, while the uptake of [18F]flumazenil was down-regulated on day 14. When comparing the left (contralateral) and right (LFPI) hemispheres, the differences between the two in neuroinflammation were obvious. Our results demonstrate a potential way to measure the molecular alterations in a rodent-based TBI model using PET imaging with [11C]PBR28 and [18F]flumazenil. These radioligands are promising options that can be eventually used in exploring the complex in vivo pharmacokinetics and delivery mechanisms of nanoparticles in TBI treatment.
Collapse
Affiliation(s)
- Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Cognitive Neuroimaging Centre, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
- Correspondence: (P.P.); (B.G.); Tel.:+65-69041186 (P.P.)
| | - Chang-Tong Yang
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Zhimin Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
| | - Mathangi Palanivel
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
| | - Kian Chye Ng
- DSO National Laboratories (Kent Ridge), 27 Medical Drive, Singapore 117510, Singapore; (K.C.N.); (J.L.)
| | - Jia Lu
- DSO National Laboratories (Kent Ridge), 27 Medical Drive, Singapore 117510, Singapore; (K.C.N.); (J.L.)
| | - Jan Carlstedt-Duke
- President’s Office, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore;
| | - Christer Halldin
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Department of Clinical Neuroscience, Karolinska Institute, S-171 76 Stockholm, Sweden
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (C.-T.Y.); (Z.W.); (M.P.); (C.H.)
- Cognitive Neuroimaging Centre, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, S-171 76 Stockholm, Sweden
- Correspondence: (P.P.); (B.G.); Tel.:+65-69041186 (P.P.)
| |
Collapse
|
39
|
|
40
|
Kreisl WC, Kim MJ, Coughlin JM, Henter ID, Owen DR, Innis RB. PET imaging of neuroinflammation in neurological disorders. Lancet Neurol 2020; 19:940-950. [PMID: 33098803 PMCID: PMC7912433 DOI: 10.1016/s1474-4422(20)30346-x] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/06/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022]
Abstract
A growing need exists for reliable in-vivo measurement of neuroinflammation to better characterise the inflammatory processes underlying various diseases and to inform the development of novel therapeutics that target deleterious glial activity. PET is well suited to quantify neuroinflammation and has the potential to discriminate components of the neuroimmune response. However, there are several obstacles to the reliable quantification of neuroinflammation by PET imaging. Despite these challenges, PET studies have consistently identified associations between neuroimmune responses and pathophysiology in brain disorders such as Alzheimer's disease. Tissue studies have also begun to clarify the meaning of changes in PET signal in some diseases. Furthermore, although PET imaging of neuroinflammation does not have an established clinical application, novel targets are under investigation and a small but growing number of studies have suggested that this imaging modality could have a role in drug development. Future studies are needed to further improve our knowledge of the cellular mechanisms that underlie changes in PET signal, how immune response contributes to neurological disease, and how it might be therapeutically modified.
Collapse
Affiliation(s)
- William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jennifer M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
41
|
Downer OM, Marcus RE, Zürcher NR, Hooker JM. Tracing the History of the Human Translocator Protein to Recent Neurodegenerative and Psychiatric Imaging. ACS Chem Neurosci 2020; 11:2192-2200. [PMID: 32662626 DOI: 10.1021/acschemneuro.0c00362] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The human 18 kDa translocator protein (TSPO) has been widely used as a measure of glial activation in health and disease. With the continuous progress of radiotracers with increased affinity and selectivity, associations between TSPO expression, disease severity, and progression have been examined, particularly in neurodegenerative disorders such as multiple sclerosis (MS), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). However, findings in psychiatric disorders have prompted reassessment of the interpretation of regional TSPO expression differences in the brain, specifically with respect to potential neuroinflammatory components. This "mini" Review aims to guide readers through the complexity of TSPO imaging research by identifying the successes, challenges, and promising new directions of the field. We will provide a brief history of how TSPO imaging has evolved over the last three decades and present lessons learned in the context of neurodegenerative and psychiatric disorders.
Collapse
Affiliation(s)
- Olivia M. Downer
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
| | - Rachel E.G. Marcus
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
| | - Nicole R. Zürcher
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
- Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Jacob M. Hooker
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
- Harvard Medical School, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
42
|
Sridharan S, Raffel J, Nandoskar A, Record C, Brooks DJ, Owen D, Sharp D, Muraro PA, Gunn R, Nicholas R. Confirmation of Specific Binding of the 18-kDa Translocator Protein (TSPO) Radioligand [ 18F]GE-180: a Blocking Study Using XBD173 in Multiple Sclerosis Normal Appearing White and Grey Matter. Mol Imaging Biol 2020; 21:935-944. [PMID: 30796709 DOI: 10.1007/s11307-019-01323-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Measurements of non-displaceable binding (VND) of positron emission tomography (PET) ligands are not often made in vivo in humans because they require ligands to displace binding to target receptors and there are few readily available, safe ones to use. A technique to measure VND for ligands for the 18-kDa translocator protein (TSPO) has recently been developed which compares the total volume of distribution (VT) before and after administration of the TSPO ligand XBD173. Here, we used XBD173 with an occupancy plot to quantify VND for two TSPO radiotracers, [18F]GE-180 and [11C]PBR28, in cohorts of people with multiple sclerosis (MS). Additionally, we compared plots of subjects carrying high (HAB) or mixed binding (MAB) affinity polymorphisms of TSPO to estimate VND without receptor blockade. PROCEDURES Twelve people with MS underwent baseline MRI and 90-min dynamic [18F]GE-180 PET or [11C]PBR28 PET (n = 6; three HAB, three MAB each). Arterial blood sampling was used to generate plasma input functions for the two-tissue compartment model. VND was calculated using two independent methods: the occupancy plot (by modelling the differences in signal post XBD173) and the polymorphism plot (by modelling the differences in signal across presence and absence of rs6971 genotypes). RESULTS Whole brain VT (mean ± standard deviation) was 0.29 ± 0.17 ml/cm3 for [18F]GE-180 and 5.01 ± 1.88 ml/cm3 for [11C]PBR28. Using the occupancy and polymorphism plots respectively, VND for [18F]GE-180 was 0.11 ml/cm3 (95 % CI = 0.02, 0.16) and 0.20 ml/cm3 (0.16, 0.34), accounting for, on average, 55 % of VT in the whole brain. For [11C]PBR28, these values were 3.81 ml/cm3 (3.02, 4.21) and 3.49 ml/cm3 (1.38, 4.27), accounting for 67 % of average whole brain VT. CONCLUSIONS Although VT for [18F]GE-180 is low, indicating low brain penetration, half the signal shown by MS subjects reflected specific TSPO binding. VT for [11C]PBR28 was higher and two thirds of the binding was non-specific. No brain ROIs were devoid of specific signal, further confirming that true reference tissue approaches are potentially problematic for estimating TSPO levels.
Collapse
Affiliation(s)
- Sujata Sridharan
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.
| | - Joel Raffel
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Ashwini Nandoskar
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Chris Record
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - David J Brooks
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Institute of Neuroscience, Newcastle upon Tyne University, Newcastle upon Tyne, UK
| | - David Owen
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - David Sharp
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Paolo A Muraro
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | | | - Richard Nicholas
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
43
|
Letter to the Editor re: Confirmation of Specific Binding of the 18-kDa Translocator Protein (TSPO) Radioligand [ 18F]GE-180: a Blocking Study Using XBD173 in Multiple Sclerosis Normal Appearing White and Grey Matter. Mol Imaging Biol 2020; 22:10-12. [PMID: 31641965 DOI: 10.1007/s11307-019-01433-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
44
|
Reliable quantification of 18F-GE-180 PET neuroinflammation studies using an individually scaled population-based input function or late tissue-to-blood ratio. Eur J Nucl Med Mol Imaging 2020; 47:2887-2900. [PMID: 32322915 PMCID: PMC7651670 DOI: 10.1007/s00259-020-04810-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/02/2020] [Indexed: 01/23/2023]
Abstract
Purpose Tracer kinetic modeling of tissue time activity curves and the individual input function based on arterial blood sampling and metabolite correction is the gold standard for quantitative characterization of microglia activation by PET with the translocator protein (TSPO) ligand 18F-GE-180. This study tested simplified methods for quantification of 18F-GE-180 PET. Methods Dynamic 18F-GE-180 PET with arterial blood sampling and metabolite correction was performed in five healthy volunteers and 20 liver-transplanted patients. Population-based input function templates were generated by averaging individual input functions normalized to the total area under the input function using a leave-one-out approach. Individual population-based input functions were obtained by scaling the input function template with the individual parent activity concentration of 18F-GE-180 in arterial plasma in a blood sample drawn at 27.5 min or by the individual administered tracer activity, respectively. The total 18F-GE-180 distribution volume (VT) was estimated in 12 regions-of-interest (ROIs) by the invasive Logan plot using the measured or the population-based input functions. Late ROI-to-whole-blood and ROI-to-cerebellum ratio were also computed. Results Correlation with the reference VT (with individually measured input function) was very high for VT with the population-based input function scaled with the blood sample and for the ROI-to-whole-blood ratio (Pearson correlation coefficient = 0.989 ± 0.006 and 0.970 ± 0.005). The correlation was only moderate for VT with the population-based input function scaled with tracer activity dose and for the ROI-to-cerebellum ratio (0.653 ± 0.074 and 0.384 ± 0.177). Reference VT, population-based VT with scaling by the blood sample, and ROI-to-whole-blood ratio were sensitive to the TSPO gene polymorphism. Population-based VT with scaling to the administered tracer activity and the ROI-to-cerebellum ratio failed to detect a polymorphism effect. Conclusion These results support the use of a population-based input function scaled with a single blood sample or the ROI-to-whole-blood ratio at a late time point for simplified quantitative analysis of 18F-GE-180 PET. Electronic supplementary material The online version of this article (10.1007/s00259-020-04810-1) contains supplementary material, which is available to authorized users.
Collapse
|
45
|
Zanotti-Fregonara P, Pascual B, Rostomily RC, Rizzo G, Veronese M, Masdeu JC, Turkheimer F. Anatomy of 18F-GE180, a failed radioligand for the TSPO protein. Eur J Nucl Med Mol Imaging 2020; 47:2233-2236. [DOI: 10.1007/s00259-020-04732-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 02/17/2020] [Indexed: 12/21/2022]
|
46
|
Head-to-head comparison of 11C-PBR28 and 11C-ER176 for quantification of the translocator protein in the human brain. Eur J Nucl Med Mol Imaging 2019; 46:1822-1829. [DOI: 10.1007/s00259-019-04349-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/29/2019] [Indexed: 10/26/2022]
|
47
|
Qiao L, Fisher E, McMurray L, Milicevic Sephton S, Hird M, Kuzhuppilly-Ramakrishnan N, Williamson DJ, Zhou X, Werry E, Kassiou M, Luthra S, Trigg W, Aigbirhio FI. Radiosynthesis of (R,S)-[ 18 F]GE387: A Potential PET Radiotracer for Imaging Translocator Protein 18 kDa (TSPO) with Low Binding Sensitivity to the Human Gene Polymorphism rs6971. ChemMedChem 2019; 14:982-993. [PMID: 30900397 PMCID: PMC6563049 DOI: 10.1002/cmdc.201900023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/26/2019] [Indexed: 12/14/2022]
Abstract
Translocator protein (TSPO) is a biomarker of neuroinflammation, which is a hallmark of many neurodegenerative diseases and has been exploited as a positron emission tomography (PET) target. Carbon-11-labelled PK11195 remains the most applied agent for imaging TSPO, despite its short-lived isotope and low brain permeability. Second-generation radiotracers show variance in affinity amongst subjects (low-, mixed-, and high-affinity binders) caused by the genetic polymorphism (rs6971) of the TSPO gene. To overcome these limitations, a new structural scaffold was explored based on the TSPO pharmacophore, and the analogue with a low-affinity binder/high-affinity binder (LAB/HAB) ratio similar (1.2 vs. 1.3) to that of (R)-[11 C]PK11195 was investigated. The synthesis of the reference compound was accomplished in six steps and 9 % overall yield, and the precursor was prepared in eight steps and 8 % overall yield. The chiral separation of the reference and precursor compounds was performed using supercritical fluid chromatography with >95 % ee. The absolute configuration was determined by circular dichroism. Optimisation of reaction conditions for manual radiolabelling revealed acetonitrile as a preferred solvent at 100 °C. Automation of this radiolabelling method provided R and S enantiomers in respective 21.3±16.7 and 25.6±7.1 % decay-corrected yields and molar activities of 55.8±35.6 and 63.5±39.5 GBq μmol-1 (n=3). Injection of the racemic analogue into a healthy rat confirmed passage through the blood-brain barrier.
Collapse
Affiliation(s)
- Luxi Qiao
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Emily Fisher
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Lindsay McMurray
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Selena Milicevic Sephton
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Matthew Hird
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Nisha Kuzhuppilly-Ramakrishnan
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - David J Williamson
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Xiouyun Zhou
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Eryn Werry
- School of Chemistry, The University of Sydney, Building F11, Eastern Avenue, Sydney, NSW, 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Building F11, Eastern Avenue, Sydney, NSW, 2006, Australia
| | | | | | - Franklin I Aigbirhio
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| |
Collapse
|
48
|
Zanotti-Fregonara P, Veronese M, Pascual B, Rostomily RC, Turkheimer F, Masdeu JC. The validity of 18F-GE180 as a TSPO imaging agent. Eur J Nucl Med Mol Imaging 2019; 46:1205-1207. [PMID: 30656358 DOI: 10.1007/s00259-019-4268-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/07/2019] [Indexed: 01/23/2023]
Affiliation(s)
- Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center and Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - Belen Pascual
- Nantz National Alzheimer Center and Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Robert C Rostomily
- Department of Neurosurgery, Houston Methodist Hospital and Research Institute, Houston, TX, USA
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - Joseph C Masdeu
- Nantz National Alzheimer Center and Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| |
Collapse
|
49
|
Chaney A, Williams SR, Boutin H. In vivo molecular imaging of neuroinflammation in Alzheimer's disease. J Neurochem 2018; 149:438-451. [PMID: 30339715 PMCID: PMC6563454 DOI: 10.1111/jnc.14615] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/24/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022]
Abstract
It has become increasingly evident that neuroinflammation plays a critical role in the pathophysiology of Alzheimer's disease (AD) and other neurodegenerative disorders. Increased glial cell activation is consistently reported in both rodent models of AD and in AD patients. Moreover, recent genome wide association studies have revealed multiple genes associated with inflammation and immunity are significantly associated with an increased risk of AD development (e.g. TREM2). Non‐invasive in vivo detection and tracking of neuroinflammation is necessary to enhance our understanding of the contribution of neuroinflammation to the initiation and progression of AD. Importantly, accurate methods of quantifying neuroinflammation may aid early diagnosis and serve as an output for therapeutic monitoring and disease management. This review details current in vivo imaging biomarkers of neuroinflammation being explored and summarizes both pre‐clinical and clinical results from molecular imaging studies investigating the role of neuroinflammation in AD, with a focus on positron emission tomography and magnetic resonance spectroscopy (MRS). ![]()
Collapse
Affiliation(s)
- Aisling Chaney
- School of Health Sciences, Division of Informatics, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK.,Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Steve R Williams
- School of Health Sciences, Division of Informatics, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre University of Manchester, Manchester, UK
| | - Herve Boutin
- Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK.,School of Biological Sciences, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| |
Collapse
|
50
|
Tang D, Fujinaga M, Hatori A, Zhang Y, Yamasaki T, Xie L, Mori W, Kumata K, Liu J, Manning HC, Huang G, Zhang MR. Evaluation of the novel TSPO radiotracer 2-(7-butyl-2-(4-(2-([ 18F]fluoroethoxy)phenyl)-5-methylpyrazolo[1,5-a]pyrimidin-3-yl)-N,N-diethylacetamide in a preclinical model of neuroinflammation. Eur J Med Chem 2018; 150:1-8. [PMID: 29505933 DOI: 10.1016/j.ejmech.2018.02.076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 12/13/2022]
Abstract
Translocator Protein (18 kDa, TSPO) is regarded as a useful biomarker for neuroinflammation imaging. TSPO PET imaging could be used to understand the role of neuroinflammation in brain diseases and as a tool for evaluating novel therapeutic effects. As a promising TSPO probe, [18F]DPA-714 is highly specific and offers reliable quantification of TSPO in vivo. In this study, we further radiosynthesized and evaluated another novel TSPO probe, 2-(7-butyl-2-(4-(2-[18F]fluoroethoxy)phenyl)-5-methylpyrazolo[1,5-a]pyrimidin-3-yl)-N,N-diethylacetamide ([18F]VUIIS1018A), which features a 700-fold higher binding affinity for TSPO than that of [18F]DPA-714. We evaluated the performance of [18F]VUIIS1018A using dynamic in vivo PET imaging, radiometabolite analysis, in vitro autoradiography assays, biodistribution analysis, and blocking assays. In vivo study using this probe demonstrated high signal-to-noise ratio, binding potential (BPND), and binding specificity in preclinical neuroinflammation studies. Taken together, these findings indicate that [18F]VUIIS1018A may serve as a novel TSPO PET probe for neuroinflammation imaging.
Collapse
Affiliation(s)
- Dewei Tang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China; Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Masayuki Fujinaga
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Akiko Hatori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yiding Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tomoteru Yamasaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Lin Xie
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Wakana Mori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Katsushi Kumata
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Jianjun Liu
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China; Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - H Charles Manning
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Gang Huang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Institute of Clinical Nuclear Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institute for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China.
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan.
| |
Collapse
|