1
|
Meng H, Chen X, He L, Chunyu H, Zhou Q, Wang J, Qu Q, Hai W, Zhang Y, Li B, Chen S, Zhang M. Comparison of 18F-DPA714 PET/MRI and 18F-FDG PET/CT in patients with autoimmune encephalitis: a cross-sectional study. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07305-z. [PMID: 40317304 DOI: 10.1007/s00259-025-07305-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND AND AIM The positive detection rate of MRI in autoimmune encephalitis (AIE) patients is low, often failing to make a rapid and accurate localization diagnosis. Both 18F-DPA714 and 18F-FDG PET imaging techniques have shown their advantages in the diagnosis of AIE. Therefore, the aim of this study was to conduct a head-to-head comparison of the diagnostic value between 18F-DPA714 and 18F-FDG PET in patients with AIE. METHODS This cross-sectional study included 23 antibody-positive and 2 possible antibody-negative AIE patients, along with 10 healthy controls. All patients underwent sequential 18F-DPA714 PET/MRI and 18F-FDG PET/CT examinations within one week. A follow-up of 6 to 12 months after the baseline examinations was performed to assess the change of patients' clinical symptoms. Positive findings for 18F-DPA714 PET were defined as Z-score above 2.0 in the corresponding brain regions of healthy controls. Positive results for 18F-FDG PET were indicated by the Z-score above 2.0 or below -2.0 obtained after comparison with the normal population database. RESULTS Both 18F-FDG PET and 18F-DPA714 PET showed higher positivity rates than MRI (81% vs 44%, P = 0.006 and 76% vs 44%, P = 0.008, respectively). A statistically significant association was found between the uptake intensity and extent of 18F-DPA714 but not 18F-FDG with the mRS scores and the CASE scores. Patients with epilepsy showed a significantly higher uptake in the whole-cortex of both 18F-DPA714 (P = 0.0014) and 18F-FDG (P = 0.0051) and in the temporal lobe of 18F-FDG (P = 0.0216) compared to those patients without. Higher 18F-DPA714 SUVR (P < 0.001) and lower 18F-FDG SUVR (P < 0.001) in the whole-cortex of patients with long-term cognitive impairment than those patients without were observed. CONCLUSION 18F-DPA714 PET showed its better correlation with disease severity of AIE than 18F-FDG PET, serving as a supplementary tool to 18F-FDG PET and MRI.
Collapse
Affiliation(s)
- Huanyu Meng
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Department of Neurology, Xinrui Hospital, Wuxi, China
| | - Xiaoyu Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Lu He
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Hangxing Chunyu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qinming Zhou
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jin Wang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Qu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Shanxi, China
| | - Sheng Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
- Department of Neurology, Xinrui Hospital, Wuxi, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Shanxi, China.
| |
Collapse
|
2
|
Zhang S, Wang X, Gao X, Chen X, Li L, Li G, Liu C, Miao Y, Wang R, Hu K. Radiopharmaceuticals and their applications in medicine. Signal Transduct Target Ther 2025; 10:1. [PMID: 39747850 PMCID: PMC11697352 DOI: 10.1038/s41392-024-02041-6] [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: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/28/2024] [Indexed: 01/04/2025] Open
Abstract
Radiopharmaceuticals involve the local delivery of radionuclides to targeted lesions for the diagnosis and treatment of multiple diseases. Radiopharmaceutical therapy, which directly causes systematic and irreparable damage to targeted cells, has attracted increasing attention in the treatment of refractory diseases that are not sensitive to current therapies. As the Food and Drug Administration (FDA) approvals of [177Lu]Lu-DOTA-TATE, [177Lu]Lu-PSMA-617 and their complementary diagnostic agents, namely, [68Ga]Ga-DOTA-TATE and [68Ga]Ga-PSMA-11, targeted radiopharmaceutical-based theranostics (radiotheranostics) are being increasingly implemented in clinical practice in oncology, which lead to a new era of radiopharmaceuticals. The new generation of radiopharmaceuticals utilizes a targeting vector to achieve the accurate delivery of radionuclides to lesions and avoid off-target deposition, making it possible to improve the efficiency and biosafety of tumour diagnosis and therapy. Numerous studies have focused on developing novel radiopharmaceuticals targeting a broader range of disease targets, demonstrating remarkable in vivo performance. These include high tumor uptake, prolonged retention time, and favorable pharmacokinetic properties that align with clinical standards. While radiotheranostics have been widely applied in tumor diagnosis and therapy, their applications are now expanding to neurodegenerative diseases, cardiovascular diseases, and inflammation. Furthermore, radiotheranostic-empowered precision medicine is revolutionizing the cancer treatment paradigm. Diagnostic radiopharmaceuticals play a pivotal role in patient stratification and treatment planning, leading to improved therapeutic outcomes in targeted radionuclide therapy. This review offers a comprehensive overview of the evolution of radiopharmaceuticals, including both FDA-approved and clinically investigated agents, and explores the mechanisms of cell death induced by radiopharmaceuticals. It emphasizes the significance and future prospects of theranostic-based radiopharmaceuticals in advancing precision medicine.
Collapse
Grants
- 82372002 National Natural Science Foundation of China (National Science Foundation of China)
- 0104002 Beijing Nova Program
- L248087; L234044 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Nonprofit Central Research Institute Fund of the Chinese Academy of Medical Sciences (No. 2022-RC350-04), the CAMS Innovation Fund for Medical Sciences (Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001), the National Key Research and Development Program of China (No. 2022YFE0111700),the Fundamental Research Funds for the Central Universities (Nos. 3332023044 and 3332023151), the CIRP Open Fund of Radiation Protection Laboratories (No. ZHYLYB2021005), and the China National Nuclear Corporation Young Talent Program.
- Fundamental Research Funds for the Central Universities,Nos. 3332023044
- Fundamental Research Funds for the Central Universities,Nos. 3332023151
- he Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,No. 2022-RC350-04;the CAMS Innovation Fund for Medical Sciences,Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001;the National Key Research and Development Program of China,No. 2022YFE0111700
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Linger Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Yuan Miao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 2019RU066, 730000, Lanzhou, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
3
|
Meng H, He L, Chunyu H, Zhou Q, Wang J, Qu Q, Hai W, Zhang Y, Li B, Zhang M, Chen S. 18F-DPA714 PET/MRI as a potential imaging tool for detecting possible antibody-negative autoimmune encephalitis: a prospective study. J Neurol 2024; 271:7592-7604. [PMID: 39294471 DOI: 10.1007/s00415-024-12690-w] [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: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND AND OBJECTIVES Conventional magnetic resonance imaging (MRI) used for detecting possible antibody-negative autoimmune encephalitis (AIE) often fails to meet the diagnostic requirements of this disease. Positron emission tomography (PET) with a translocator protein radioligand can help visualize microglia distribution density in inflammation-related diseases, thereby offering potentially incremental value to conventional MRI for the in vivo assessment of possible antibody-negative AIE. METHODS In this prospective study, 15 participants diagnosed with possible antibody-negative AIE and 10 healthy controls were enrolled (ClinicalTrials.gov: NCT05293405, dated March 15, 2022). All participants underwent hybrid 18F-DPA714 PET/MRI and evaluation for modified Rankin scale (mRS) score, clinical assessment scale for AIE (CASE), and appropriate antibodies. A positive finding was defined as the intensity of 18F-DPA714 uptake that was above a threshold of mean standardized uptake value ratio (SUVR) + two standard deviations of SUVR within the corresponding brain regions of healthy controls. RESULTS The positive detection rate of 18F-DPA714 PET for possible antibody-negative AIE was significantly higher than that of brain MRI (10/15 [67%] vs. 3/15 [20%]; P = 0.039). In addition, both the intensity and extent of 18F-DPA714 uptake were significantly associated with the CASE score (P = 0.002 and 0.001). Meanwhile, SUVR levels in the cerebellar region were significantly higher in patients with ataxia than in those without ataxia (P = 0.006). Furthermore, 18F-DPA714 uptake decreased in 5/10 [50%] patients who underwent follow-up PET/MRI, which mirrored their symptom relief. CONCLUSION 18F-DPA714 PET demonstrated its potentially incremental value to conventional MRI for detecting possible antibody-negative AIE.
Collapse
Affiliation(s)
- Huanyu Meng
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Lu He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Hangxing Chunyu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qinming Zhou
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jin Wang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Qu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanxi Medical University-Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi, China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanxi Medical University-Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi, China.
| | - Sheng Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
- Department of Neurology, Xinrui Hospital, Wuxi, China.
| |
Collapse
|
4
|
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
|
5
|
Qin L, Xiao L, Zhu H, Du Y, Tang Y, Feng L. Translocator protein (18 kDa) positron emission tomography imaging as a biomarker of neuroinflammation in epilepsy. Neurol Sci 2024; 45:5201-5211. [PMID: 38879831 DOI: 10.1007/s10072-024-07648-9] [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: 03/14/2024] [Accepted: 06/11/2024] [Indexed: 10/13/2024]
Abstract
Increasing evidence indicate that neuroinflammation triggered by glial cells plays a significant role in epileptogenesis. To this effect, the overexpression of translocator protein 18 kDa (TSPO) in activated microglia and astrocytes has been identified as an inflammatory biomarker in epilepsy. It is now possible to quantify neuroinflammation using non-invasive positron emission tomography (PET) imaging of TSPO. With the advancement of radiotracers, TSPO PET has become an innovative tool in elucidating the "neuroinflammatory machinery" of drug-resistant epilepsy. Furthermore, TSPO PET has demonstrated potential in detecting MRI-negative epileptogenic zones (EZ) and provided an innovative perspective in epileptic medical treatment. This manuscript presents a comprehensive exploration of the neuroinflammatory mechanisms of epilepsy, alongside a thorough review of TSPO PET studies conducted in clinical and preclinical settings. The primary objective is to deepen our understanding of epilepsy progression and to establish TSPO PET as an effective monitoring tool for treatment efficacy.
Collapse
Affiliation(s)
- Li Qin
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Haoyue Zhu
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yangsa Du
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Li Feng
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Neurology, Xiangya Hospital, Central South University (Jiangxi Branch), Nanchang, 330000, Jiangxi, China.
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 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
|
7
|
Bagnato F, Sati P, Hemond CC, Elliott C, Gauthier SA, Harrison DM, Mainero C, Oh J, Pitt D, Shinohara RT, Smith SA, Trapp B, Azevedo CJ, Calabresi PA, Henry RG, Laule C, Ontaneda D, Rooney WD, Sicotte NL, Reich DS, Absinta M. Imaging chronic active lesions in multiple sclerosis: a consensus statement. Brain 2024; 147:2913-2933. [PMID: 38226694 PMCID: PMC11370808 DOI: 10.1093/brain/awae013] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 01/17/2024] Open
Abstract
Chronic active lesions (CAL) are an important manifestation of chronic inflammation in multiple sclerosis and have implications for non-relapsing biological progression. In recent years, the discovery of innovative MRI and PET-derived biomarkers has made it possible to detect CAL, and to some extent quantify them, in the brain of persons with multiple sclerosis, in vivo. Paramagnetic rim lesions on susceptibility-sensitive MRI sequences, MRI-defined slowly expanding lesions on T1-weighted and T2-weighted scans, and 18-kDa translocator protein-positive lesions on PET are promising candidate biomarkers of CAL. While partially overlapping, these biomarkers do not have equivalent sensitivity and specificity to histopathological CAL. Standardization in the use of available imaging measures for CAL identification, quantification and monitoring is lacking. To fast-forward clinical translation of CAL, the North American Imaging in Multiple Sclerosis Cooperative developed a consensus statement, which provides guidance for the radiological definition and measurement of CAL. The proposed manuscript presents this consensus statement, summarizes the multistep process leading to it, and identifies the remaining major gaps in knowledge.
Collapse
Affiliation(s)
- Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Department of Neurology, Nashville VA Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Pascal Sati
- Neuroimaging Program, Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Christopher C Hemond
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | | | - Susan A Gauthier
- Department of Neurology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Neurology, Baltimore VA Medical Center, VA Maryland Healthcare System, Baltimore, MD 21201, USA
| | - Caterina Mainero
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jiwon Oh
- Division of Neurology, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S, Canada
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Russell T Shinohara
- Penn Statistics in Imaging and Visualization Endeavor, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Biomedical Image Computing and Analytics, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seth A Smith
- Department of Radiology and Radiological Sciences, Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Bruce Trapp
- Department on Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Christina J Azevedo
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90007, USA
| | - Peter A Calabresi
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Roland G Henry
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Cornelia Laule
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH 44195, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR 97239, USA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Martina Absinta
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Translational Neuropathology Unit, Division of Neuroscience, Institute of Experimental Neurology, Vita-Salute San Raffaele University and IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| |
Collapse
|
8
|
Vinnakota JM, Biavasco F, Schwabenland M, Chhatbar C, Adams RC, Erny D, Duquesne S, El Khawanky N, Schmidt D, Fetsch V, Zähringer A, Salié H, Athanassopoulos D, Braun LM, Javorniczky NR, Ho JNHG, Kierdorf K, Marks R, Wäsch R, Simonetta F, Andrieux G, Pfeifer D, Monaco G, Capitini C, Fry TJ, Blank T, Blazar BR, Wagner E, Theobald M, Sommer C, Stelljes M, Reicherts C, Jeibmann A, Schittenhelm J, Monoranu CM, Rosenwald A, Kortüm M, Rasche L, Einsele H, Meyer PT, Brumberg J, Völkl S, Mackensen A, Coras R, von Bergwelt-Baildon M, Albert NL, Bartos LM, Brendel M, Holzgreve A, Mack M, Boerries M, Mackall CL, Duyster J, Henneke P, Priller J, Köhler N, Strübing F, Bengsch B, Ruella M, Subklewe M, von Baumgarten L, Gill S, Prinz M, Zeiser R. Targeting TGFβ-activated kinase-1 activation in microglia reduces CAR T immune effector cell-associated neurotoxicity syndrome. NATURE CANCER 2024; 5:1227-1249. [PMID: 38741011 DOI: 10.1038/s43018-024-00764-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 03/27/2024] [Indexed: 05/16/2024]
Abstract
Cancer immunotherapy with chimeric antigen receptor (CAR) T cells can cause immune effector cell-associated neurotoxicity syndrome (ICANS). However, the molecular mechanisms leading to ICANS are not well understood. Here we examined the role of microglia using mouse models and cohorts of individuals with ICANS. CD19-directed CAR (CAR19) T cell transfer in B cell lymphoma-bearing mice caused microglia activation and neurocognitive deficits. The TGFβ-activated kinase-1 (TAK1)-NF-κB-p38 MAPK pathway was activated in microglia after CAR19 T cell transfer. Pharmacological TAK1 inhibition or genetic Tak1 deletion in microglia using Cx3cr1CreER:Tak1fl/fl mice resulted in reduced microglia activation and improved neurocognitive activity. TAK1 inhibition allowed for potent CAR19-induced antilymphoma effects. Individuals with ICANS exhibited microglia activation in vivo when studied by translocator protein positron emission tomography, and imaging mass cytometry revealed a shift from resting to activated microglia. In summary, we prove a role for microglia in ICANS pathophysiology, identify the TAK1-NF-κB-p38 MAPK axis as a pathogenic signaling pathway and provide a rationale to test TAK1 inhibition in a clinical trial for ICANS prevention after CAR19 T cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Janaki Manoja Vinnakota
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Francesca Biavasco
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marius Schwabenland
- Institute for Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Chintan Chhatbar
- Institute for Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Rachael C Adams
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Daniel Erny
- Institute for Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Sandra Duquesne
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nadia El Khawanky
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Dominik Schmidt
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Viktor Fetsch
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Alexander Zähringer
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Henrike Salié
- Department of Medicine II, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dimitrios Athanassopoulos
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas M Braun
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Nora R Javorniczky
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jenny N H G Ho
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute for Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Reinhard Marks
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ralph Wäsch
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Federico Simonetta
- Division of Hematology, Geneva University Hospitals Geneva, Geneva, Switzerland
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gianni Monaco
- Institute for Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Single-Cell Omics Platform Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center, University of Freiburg, Freiburg, Germany
| | - Christian Capitini
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Terry J Fry
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Blank
- Institute for Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, USA
| | - Eva Wagner
- Department of Hematology and Medical Oncology, Johannes Gutenberg-University Medical Center, Mainz, Germany
| | - Matthias Theobald
- Department of Hematology and Medical Oncology, Johannes Gutenberg-University Medical Center, Mainz, Germany
| | - Clemens Sommer
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Matthias Stelljes
- Department of Medicine/Hematology and Oncology, University of Münster, Münster, Germany
| | - Christian Reicherts
- Department of Medicine/Hematology and Oncology, University of Münster, Münster, Germany
| | - Astrid Jeibmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Jens Schittenhelm
- Department of Neuropathology, Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | | | | | - Martin Kortüm
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Joachim Brumberg
- Department of Nuclear Medicine, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, Hematology/Oncology, University Hospital, Ludwig-Maximilians Universität (LMU) Munich, Munich, Germany
| | - Nathalie L Albert
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Mack
- Department of Nephrology, University of Regensburg, Regensburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA, USA
| | - Justus Duyster
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Division of Pediatric Infectious Diseases, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Josef Priller
- Department of Psychiatry, Technischen Universität München (TUM), Munich, Germany
| | - Natalie Köhler
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Felix Strübing
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany
| | - Bertram Bengsch
- Department of Medicine II, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Marion Subklewe
- Department of Medicine III, Hematology/Oncology, University Hospital, Ludwig-Maximilians Universität (LMU) Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Louisa von Baumgarten
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Neuro-Oncology, Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Saar Gill
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Prinz
- Institute for Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Signalling Research Centres BIOSS and Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
9
|
Galldiks N, Kaufmann TJ, Vollmuth P, Lohmann P, Smits M, Veronesi MC, Langen KJ, Rudà R, Albert NL, Hattingen E, Law I, Hutterer M, Soffietti R, Vogelbaum MA, Wen PY, Weller M, Tonn JC. Challenges, limitations, and pitfalls of PET and advanced MRI in patients with brain tumors: A report of the PET/RANO group. Neuro Oncol 2024; 26:1181-1194. [PMID: 38466087 PMCID: PMC11226881 DOI: 10.1093/neuonc/noae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Indexed: 03/12/2024] Open
Abstract
Brain tumor diagnostics have significantly evolved with the use of positron emission tomography (PET) and advanced magnetic resonance imaging (MRI) techniques. In addition to anatomical MRI, these modalities may provide valuable information for several clinical applications such as differential diagnosis, delineation of tumor extent, prognostication, differentiation between tumor relapse and treatment-related changes, and the evaluation of response to anticancer therapy. In particular, joint recommendations of the Response Assessment in Neuro-Oncology (RANO) Group, the European Association of Neuro-oncology, and major European and American Nuclear Medicine societies highlighted that the additional clinical value of radiolabeled amino acids compared to anatomical MRI alone is outstanding and that its widespread clinical use should be supported. For advanced MRI and its steadily increasing use in clinical practice, the Standardization Subcommittee of the Jumpstarting Brain Tumor Drug Development Coalition provided more recently an updated acquisition protocol for the widely used dynamic susceptibility contrast perfusion MRI. Besides amino acid PET and perfusion MRI, other PET tracers and advanced MRI techniques (e.g. MR spectroscopy) are of considerable clinical interest and are increasingly integrated into everyday clinical practice. Nevertheless, these modalities have shortcomings which should be considered in clinical routine. This comprehensive review provides an overview of potential challenges, limitations, and pitfalls associated with PET imaging and advanced MRI techniques in patients with gliomas or brain metastases. Despite these issues, PET imaging and advanced MRI techniques continue to play an indispensable role in brain tumor management. Acknowledging and mitigating these challenges through interdisciplinary collaboration, standardized protocols, and continuous innovation will further enhance the utility of these modalities in guiding optimal patient care.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
| | | | - Philipp Vollmuth
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
| | - Marion Smits
- Department of Radiology and Nuclear Medicine and Brain Tumour Center, Erasmus MC, Rotterdam, The Netherlands
| | - Michael C Veronesi
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilians-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Hattingen
- Goethe University, Department of Neuroradiology, University Hospital Frankfurt, Frankfurt, Germany
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Markus Hutterer
- Department of Neurology with Acute Geriatrics, Saint John of God Hospital, Linz, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Michael A Vogelbaum
- Department of Neuro-Oncology and Neurosurgery, Moffit Cancer Center, Tampa, Florida, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, and University Hospital of Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Joerg-Christian Tonn
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
10
|
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
|
11
|
Knyzeliene A, MacAskill MG, Alcaide-Corral CJ, Morgan TEF, Henry MC, Lucatelli C, Pimlott SL, Sutherland A, Tavares AAS. [ 18F]LW223 has low non-displaceable binding in murine brain, enabling high sensitivity TSPO PET imaging. J Cereb Blood Flow Metab 2024; 44:397-406. [PMID: 37795635 PMCID: PMC10870961 DOI: 10.1177/0271678x231205661] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/01/2023] [Accepted: 09/09/2023] [Indexed: 10/06/2023]
Abstract
Neuroinflammation is associated with a number of brain diseases, making it a common feature of cerebral pathology. Among the best-known biomarkers for neuroinflammation in Positron Emission Tomography (PET) research is the 18 kDa translocator protein (TSPO). This study aims to investigate the binding kinetics of a novel TSPO PET radiotracer, [18F]LW223, in mice and specifically assess its volume of non-displaceable binding (VND) in brain as well as investigate the use of simplified analysis approaches for quantification of [18F]LW223 PET data. Adult male mice were injected with [18F]LW223 and varying concentrations of LW223 (0.003-0.55 mg/kg) to estimate VND of [18F]LW223. Dynamic PET imaging with arterial input function studies and radiometabolite studies were conducted. Simplified quantification methods, standard uptake values (SUV) and apparent volume of distribution (VTapp), were investigated. [18F]LW223 had low VND in the brain (<10% of total binding) and low radiometabolism (∼15-20%). The 2-tissue compartment model provided the best fit for [18F]LW223 PET data, although its correlation with SUV90-120min or VTapp allowed for [18F]LW223 brain PET data quantification in healthy animals while using simpler experimental and analytical approaches. [18F]LW223 has the required properties to become a successful TSPO PET radiotracer.
Collapse
Affiliation(s)
- Agne Knyzeliene
- BHF-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Mark G MacAskill
- BHF-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Carlos J Alcaide-Corral
- BHF-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Timaeus EF Morgan
- BHF-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | | | | | - Sally L Pimlott
- West of Scotland PET Centre, Greater Glasgow and Clyde NHS Trust, Glasgow, UK
| | | | - Adriana AS Tavares
- BHF-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Kipp M. Astrocytes: Lessons Learned from the Cuprizone Model. Int J Mol Sci 2023; 24:16420. [PMID: 38003609 PMCID: PMC10671869 DOI: 10.3390/ijms242216420] [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: 10/12/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
A diverse array of neurological and psychiatric disorders, including multiple sclerosis, Alzheimer's disease, and schizophrenia, exhibit distinct myelin abnormalities at both the molecular and histological levels. These aberrations are closely linked to dysfunction of oligodendrocytes and alterations in myelin structure, which may be pivotal factors contributing to the disconnection of brain regions and the resulting characteristic clinical impairments observed in these conditions. Astrocytes, which significantly outnumber neurons in the central nervous system by a five-to-one ratio, play indispensable roles in the development, maintenance, and overall well-being of neurons and oligodendrocytes. Consequently, they emerge as potential key players in the onset and progression of a myriad of neurological and psychiatric disorders. Furthermore, targeting astrocytes represents a promising avenue for therapeutic intervention in such disorders. To gain deeper insights into the functions of astrocytes in the context of myelin-related disorders, it is imperative to employ appropriate in vivo models that faithfully recapitulate specific aspects of complex human diseases in a reliable and reproducible manner. One such model is the cuprizone model, wherein metabolic dysfunction in oligodendrocytes initiates an early response involving microglia and astrocyte activation, culminating in multifocal demyelination. Remarkably, following the cessation of cuprizone intoxication, a spontaneous process of endogenous remyelination occurs. In this review article, we provide a historical overview of studies investigating the responses and putative functions of astrocytes in the cuprizone model. Following that, we list previously published works that illuminate various aspects of the biology and function of astrocytes in this multiple sclerosis model. Some of the studies are discussed in more detail in the context of astrocyte biology and pathology. Our objective is twofold: to provide an invaluable overview of this burgeoning field, and, more importantly, to inspire fellow researchers to embark on experimental investigations to elucidate the multifaceted functions of this pivotal glial cell subpopulation.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
13
|
Kolabas ZI, Kuemmerle LB, Perneczky R, Förstera B, Ulukaya S, Ali M, Kapoor S, Bartos LM, Büttner M, Caliskan OS, Rong Z, Mai H, Höher L, Jeridi D, Molbay M, Khalin I, Deligiannis IK, Negwer M, Roberts K, Simats A, Carofiglio O, Todorov MI, Horvath I, Ozturk F, Hummel S, Biechele G, Zatcepin A, Unterrainer M, Gnörich J, Roodselaar J, Shrouder J, Khosravani P, Tast B, Richter L, Díaz-Marugán L, Kaltenecker D, Lux L, Chen Y, Zhao S, Rauchmann BS, Sterr M, Kunze I, Stanic K, Kan VWY, Besson-Girard S, Katzdobler S, Palleis C, Schädler J, Paetzold JC, Liebscher S, Hauser AE, Gokce O, Lickert H, Steinke H, Benakis C, Braun C, Martinez-Jimenez CP, Buerger K, Albert NL, Höglinger G, Levin J, Haass C, Kopczak A, Dichgans M, Havla J, Kümpfel T, Kerschensteiner M, Schifferer M, Simons M, Liesz A, Krahmer N, Bayraktar OA, Franzmeier N, Plesnila N, Erener S, Puelles VG, Delbridge C, Bhatia HS, Hellal F, Elsner M, Bechmann I, Ondruschka B, Brendel M, Theis FJ, Erturk A. Distinct molecular profiles of skull bone marrow in health and neurological disorders. Cell 2023; 186:3706-3725.e29. [PMID: 37562402 PMCID: PMC10443631 DOI: 10.1016/j.cell.2023.07.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/24/2023] [Accepted: 07/07/2023] [Indexed: 08/12/2023]
Abstract
The bone marrow in the skull is important for shaping immune responses in the brain and meninges, but its molecular makeup among bones and relevance in human diseases remain unclear. Here, we show that the mouse skull has the most distinct transcriptomic profile compared with other bones in states of health and injury, characterized by a late-stage neutrophil phenotype. In humans, proteome analysis reveals that the skull marrow is the most distinct, with differentially expressed neutrophil-related pathways and a unique synaptic protein signature. 3D imaging demonstrates the structural and cellular details of human skull-meninges connections (SMCs) compared with veins. Last, using translocator protein positron emission tomography (TSPO-PET) imaging, we show that the skull bone marrow reflects inflammatory brain responses with a disease-specific spatial distribution in patients with various neurological disorders. The unique molecular profile and anatomical and functional connections of the skull show its potential as a site for diagnosing, monitoring, and treating brain diseases.
Collapse
Affiliation(s)
- Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Louis B Kuemmerle
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Robert Perneczky
- Division of Mental Health in Older Adults and Alzheimer Therapy and Research Center, Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University Munich, 80336 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Benjamin Förstera
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Mayar Ali
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Saketh Kapoor
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Maren Büttner
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ozum Sehnaz Caliskan
- Institute for Diabetes and Obesity, Helmholtz Center Munich and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Zhouyi Rong
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Medical Research School (MMRS), 80336 Munich, Germany
| | - Hongcheng Mai
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Medical Research School (MMRS), 80336 Munich, Germany
| | - Luciano Höher
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Denise Jeridi
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Muge Molbay
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | | | - Moritz Negwer
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | | | - Alba Simats
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Olga Carofiglio
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Mihail I Todorov
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, 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
| | - Furkan Ozturk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Selina Hummel
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Artem Zatcepin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jay Roodselaar
- Charité - Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Joshua Shrouder
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Pardis Khosravani
- Biomedical Center (BMC), Core Facility Flow Cytometry, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Benjamin Tast
- Biomedical Center (BMC), Core Facility Flow Cytometry, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lisa Richter
- Biomedical Center (BMC), Core Facility Flow Cytometry, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Laura Díaz-Marugán
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Doris Kaltenecker
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Diabetes and Cancer, Helmholtz Munich, Munich, Germany
| | - Laurin Lux
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Ying Chen
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Shan Zhao
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Division of Mental Health in Older Adults and Alzheimer Therapy and Research Center, Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University Munich, 80336 Munich, Germany; Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK; Institute of Neuroradiology, University Hospital LMU, Munich, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ines Kunze
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Karen Stanic
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Vanessa W Y Kan
- Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany
| | - Simon Besson-Girard
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Sabrina Katzdobler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes C Paetzold
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Department of Computing, Imperial College London, London, UK
| | - Sabine Liebscher
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Anja E Hauser
- Charité - Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Hanno Steinke
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany
| | - Corinne Benakis
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Christian Braun
- Institute of Legal Medicine, Faculty of Medicine, LMU Munich, Germany
| | - Celia P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anna Kopczak
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Martin Kerschensteiner
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Center Munich and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | | | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Suheda Erener
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Claire Delbridge
- Institute of Pathology, Department of Neuropathology, Technical University Munich, TUM School of Medicine, Munich, Germany
| | - Harsharan Singh Bhatia
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Farida Hellal
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Markus Elsner
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Department of Mathematics, Technische Universität München, Garching bei München, Germany
| | - Ali Erturk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
14
|
Kim SJW, Lupo JM, Chen Y, Pampaloni MH, VanBrocklin HF, Narvid J, Kim H, Seo Y. A feasibility study for quantitative assessment of cerebrovascular malformations using flutriciclamide ([18F]GE-180) PET/MRI. Front Med (Lausanne) 2023; 10:1091463. [PMID: 37089589 PMCID: PMC10116613 DOI: 10.3389/fmed.2023.1091463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
AimNeuroinflammation plays a key role in both the pathogenesis and the progression of cerebral cavernous malformations (CCM). Flutriciclamide ([18F]GE-180) is a translocator protein (TSPO) targeting positron emission tomography (PET) tracer, developed for imaging neuroinflammation. The objectives of this study were to describe characteristics of flutriciclamide uptake in different brain tissue regions in CCM patients compared to controls, and to evaluate flutriciclamide uptake and iron deposition within CCM lesions.Materials and methodsFive patients with CCM and six controls underwent a 60 or 90 min continuous PET/MRI scan following 315 ± 68.9 MBq flutriciclamide administration. Standardized uptake value (SUV) and standardized uptake value ratio (SUVr) were obtained using the striatum as a pseudo-reference. Quantitative susceptibility maps (QSM) were used to define the location of the vascular malformation and calculate the amount of iron deposition in each lesion.ResultsIncreased flutriciclamide uptake was observed in all CCM lesions. The temporal pole demonstrated the highest radiotracer uptake; the paracentral lobule, cuneus and hippocampus exhibited moderate uptake; while the striatum had the lowest uptake, with average SUVs of 0.66, 0.55, 0.63, 0.55, and 0.33 for patient with CCM and 0.57, 0.50, 0.48, 0.42, and 0.32 for controls, respectively. Regional SUVr showed similar trends. The average SUV and QSM values in CCM lesions were 0.58 ± 0.23 g/ml and 0.30 ± 0.10 ppm. SUVs and QSM were positively correlated in CCM lesions (r = 0.53, p = 0.03).ConclusionThe distribution of flutriciclamide ([18F]GE-180) in the human brain and CCM lesions demonstrated the potential of this TSPO PET tracer as a marker of neuroinflammation that may be relevant for characterizing CCM disease progression along with QSM.
Collapse
Affiliation(s)
- Sally Ji Who Kim
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Sally Ji Who Kim,
| | - Janine M. Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Yicheng Chen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Miguel H. Pampaloni
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Jared Narvid
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, United States
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
15
|
PET Imaging of Neuro-Inflammation with Tracers Targeting the Translocator Protein (TSPO), a Systematic Review: From Bench to Bedside. Diagnostics (Basel) 2023; 13:diagnostics13061029. [PMID: 36980337 PMCID: PMC10047854 DOI: 10.3390/diagnostics13061029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Parkinson’s disease is the second most common neurodegenerative disorder, affecting 2–3% of the population of patients >65 years. Although the standard diagnosis of PD is clinical, neuroimaging plays a key role in the evaluation of patients who present symptoms related to neurodegenerative disorders. MRI, DAT-SPECT, and PET with [18F]-FDG are routinely used in the diagnosis and focus on the investigation of morphological changes, nigrostriatal degeneration or shifts in glucose metabolism in patients with parkinsonian syndromes. The aim of this study is to review the current PET radiotracers targeting TSPO, a transmembrane protein that is overexpressed by microglia in another pathophysiological process associated with neurodegenerative disorders known as neuroinflammation. To the best of our knowledge, neuroinflammation is present not only in PD but in many other neurodegenerative disorders, including AD, DLB, and MSA, as well as atypical parkinsonian syndromes. Therefore, in this study, specific patterns of microglial activation in PD and the differences in distribution volumes of these radiotracers in patients with PD as compared to other neurodegenerative disorders are reviewed.
Collapse
|
16
|
Haider A, Elghazawy NH, Dawoud A, Gebhard C, Wichmann T, Sippl W, Hoener M, Arenas E, Liang SH. Translational molecular imaging and drug development in Parkinson's disease. Mol Neurodegener 2023; 18:11. [PMID: 36759912 PMCID: PMC9912681 DOI: 10.1186/s13024-023-00600-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects elderly people and constitutes a major source of disability worldwide. Notably, the neuropathological hallmarks of PD include nigrostriatal loss and the formation of intracellular inclusion bodies containing misfolded α-synuclein protein aggregates. Cardinal motor symptoms, which include tremor, rigidity and bradykinesia, can effectively be managed with dopaminergic therapy for years following symptom onset. Nonetheless, patients ultimately develop symptoms that no longer fully respond to dopaminergic treatment. Attempts to discover disease-modifying agents have increasingly been supported by translational molecular imaging concepts, targeting the most prominent pathological hallmark of PD, α-synuclein accumulation, as well as other molecular pathways that contribute to the pathophysiology of PD. Indeed, molecular imaging modalities such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can be leveraged to study parkinsonism not only in animal models but also in living patients. For instance, mitochondrial dysfunction can be assessed with probes that target the mitochondrial complex I (MC-I), while nigrostriatal degeneration is typically evaluated with probes designed to non-invasively quantify dopaminergic nerve loss. In addition to dopaminergic imaging, serotonin transporter and N-methyl-D-aspartate (NMDA) receptor probes are increasingly used as research tools to better understand the complexity of neurotransmitter dysregulation in PD. Non-invasive quantification of neuroinflammatory processes is mainly conducted by targeting the translocator protein 18 kDa (TSPO) on activated microglia using established imaging agents. Despite the overwhelming involvement of the brain and brainstem, the pathophysiology of PD is not restricted to the central nervous system (CNS). In fact, PD also affects various peripheral organs such as the heart and gastrointestinal tract - primarily via autonomic dysfunction. As such, research into peripheral biomarkers has taken advantage of cardiac autonomic denervation in PD, allowing the differential diagnosis between PD and multiple system atrophy with probes that visualize sympathetic nerve terminals in the myocardium. Further, α-synuclein has recently gained attention as a potential peripheral biomarker in PD. This review discusses breakthrough discoveries that have led to the contemporary molecular concepts of PD pathophysiology and how they can be harnessed to develop effective imaging probes and therapeutic agents. Further, we will shed light on potential future trends, thereby focusing on potential novel diagnostic tracers and disease-modifying therapeutic interventions.
Collapse
Affiliation(s)
- Achi Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| | - Nehal H Elghazawy
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas Wichmann
- Department of Neurology/School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120, Halle, Germany
| | - Marius Hoener
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Ernest Arenas
- Karolinska Institutet, MBB, Molecular Neurobiology, Stockholm, Sweden
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
17
|
TSPO PET signal using [ 18F]GE180 is associated with survival in recurrent gliomas. Eur J Nucl Med Mol Imaging 2023; 50:859-869. [PMID: 36329288 PMCID: PMC9852133 DOI: 10.1007/s00259-022-06006-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Glioma patients, especially recurrent glioma, suffer from a poor prognosis. While advances to classify glioma on a molecular level improved prognostication at initial diagnosis, markers to prognosticate survival in the recurrent situation are still needed. As 18 kDa translocator protein (TSPO) was previously reported to be associated with aggressive histopathological glioma features, we correlated the TSPO positron emission tomography (PET) signal using [18F]GE180 in a large cohort of recurrent glioma patients with their clinical outcome. METHODS In patients with [18F]GE180 PET at glioma recurrence, [18F]GE180 PET parameters (e.g., SUVmax) as well as other imaging features (e.g., MRI volume, [18F]FET PET parameters when available) were evaluated together with patient characteristics (age, sex, Karnofsky-Performance score) and neuropathological features (e.g. WHO 2021 grade, IDH-mutation status). Uni- and multivariate Cox regression and Kaplan-Meier survival analyses were performed to identify prognostic factors for post-recurrence survival (PRS) and time to treatment failure (TTF). RESULTS Eighty-eight consecutive patients were evaluated. TSPO tracer uptake correlated with tumor grade at recurrence (p < 0.05), with no significant differences in IDH-wild-type versus IDH-mutant tumors. Within the subgroup of IDH-mutant glioma (n = 46), patients with low SUVmax (median split, ≤ 1.60) had a significantly longer PRS (median 41.6 vs. 25.3 months, p = 0.031) and TTF (32.2 vs 8.7 months, p = 0.001). Also among IDH-wild-type glioblastoma (n = 42), patients with low SUVmax (≤ 1.89) had a significantly longer PRS (median not reached vs 8.2 months, p = 0.002). SUVmax remained an independent prognostic factor for PRS in the multivariate analysis including CNS WHO 2021 grade, IDH status, and age. Tumor volume defined by [18F]FET PET or contrast-enhanced MRI correlated weakly with TSPO tracer uptake. Treatment regimen did not differ among the median split subgroups. CONCLUSION Our data suggest that TSPO PET using [18F]GE180 can help to prognosticate recurrent glioma patients even among homogeneous molecular subgroups and may therefore serve as valuable non-invasive biomarker for individualized patient management.
Collapse
|
18
|
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
|
19
|
Lee SY, Oh HR, Kim YH, Bae SH, Lee Y, Lee YS, Lee BC, Cheon GJ, Kang KW, Youn H. Cerenkov luminescence imaging of interscapular brown adipose tissue using a TSPO-targeting PET probe in the UCP1 ThermoMouse. Am J Cancer Res 2022; 12:6380-6394. [PMID: 36168637 PMCID: PMC9475450 DOI: 10.7150/thno.74828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/20/2022] [Indexed: 11/21/2022] Open
Abstract
Rationale: [18F]fluorodeoxyglucose-positron emission tomography ([18F]FDG-PET) has been widely used as an imaging technique to measure interscapular brown adipose tissue (iBAT) activity. However, it is challenging to obtain iBAT-specific images using [18F]FDG-PET because increased uptake of [18F]FDG is observed in tumors, muscle, and inflamed tissues. Uncoupling protein 1 (UCP1) in the mitochondrial membrane, a well-known molecular marker of BAT, has been proposed as a useful BAT imaging marker. Recently, the UCP1 ThermoMouse was developed as a reporter mouse for monitoring UCP1 expression and investigating BAT activation. In addition, Translocator protein-18 kDa (TSPO) located in the outer mitochondrial membrane is also overexpressed in BAT, suggesting that TSPO-targeting PET has potential for iBAT imaging. However, there are no studies monitoring BAT using TSPO-targeting PET probes in the UCP1 ThermoMouse. Moreover, the non-invasive Cerenkov luminescence imaging (CLI) using Cerenkov radiation from the PET probe has been proposed as an alternative option for PET as it is less expensive and user-friendly. Therefore, we selected [18F]fm-PBR28-d2 as a TSPO-targeting PET probe for iBAT imaging to evaluate the usefulness of CLI in the UCP1 ThermoMouse. Methods: UCP1 ThermoMouse was used to monitor UCP1 expression. Western blotting and immunohistochemistry were performed to measure the level of protein expression. [18F]fm-PBR28-d2 and [18F]FDG were used as radioactive probes for iBAT imaging. PET images were acquired with SimPET, and optical images were acquired with IVIS 100. Results: UCP1 ThermoMouse showed that UCP1 and TSPO expressions were correlated in iBAT. In both PET and CLI, the TSPO-targeting probe [18F]fm-PBR28-d2 was superior to [18F]FDG for acquiring iBAT images. The high molar activity of the probe was essential for CLI and PET imaging. We tested the feasibility of TSPO-targeting probe under cold exposure by imaging with TSPO-PET/CLI. Both signals of iBAT were clearly increased after cold stimulation. Under prolonged isoflurane anesthesia, TSPO-targeting images showed higher signals from iBAT in the short-term than in long-term groups. Conclusion: We demonstrated that TSPO-PET/CLI reflected UCP1 expression in iBAT imaging better than [18F]FDG-PET/CLI under the various conditions. Considering convenience and cost, TSPO-CLI could be used as an alternative TSPO-PET technique for iBAT imaging.
Collapse
Affiliation(s)
- Seok-Yong Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ho Rim Oh
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young-Hwa Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Hwan Bae
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yongseok Lee
- Cancer Imaging Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Imaging Center, Seoul National University Hospital, Seoul, Republic of Korea.,Radiation Medicine Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Byung Chul Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Center for Nanomolecular Imaging and Innovative Drug Development, Advanced Institutes of Convergence Technology, Seoul National University, Suwon, Republic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Imaging Center, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
20
|
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
|
21
|
Xie F, Wei W. [ 64Cu]Cu-ATSM: an emerging theranostic agent for cancer and neuroinflammation. Eur J Nucl Med Mol Imaging 2022; 49:3964-3972. [PMID: 35918492 DOI: 10.1007/s00259-022-05887-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
22
|
Unterrainer M, Mahler C, Schumacher AM, Ruf V, Blum B, Quach S, Brendel M, Rupprecht R, Bartenstein P, Kerschensteiner M, Kümpfel T, Albert NL. Amino Acid Uptake, Glucose Metabolism, and Neuroinflammation in John Cunningham Virus Associated Progressive Multifocal Leukoencephalopathy. Clin Nucl Med 2022; 47:543-544. [PMID: 35195584 DOI: 10.1097/rlu.0000000000004093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT A 69-year-old woman presented with progressive dysarthria and cognitive deficits. On MRI, a T2-hyperintense, non-contrast-enhancing lesion was found in the left precentral area. 18F-FET and 18F-FDG PET scans revealed faint amino acid uptake and glucose hypometabolism of the lesion. To assess a neuroinflammatory component, TSPO PET with 18F-GE-180 was performed, where tracer uptake markedly exceeded the T2-hyperintense areas. Histology derived from a stereotactic biopsy findings confirmed John Cunningham virus-associated progressive multifocal leukoencephalopathy. This case underlines that TSPO PET comprises distinct imaging advantages over other established radioligands such as 18F-FET and 18F-FDG in progressive multifocal leukoencephalopathy.
Collapse
Affiliation(s)
| | | | | | - Viktoria Ruf
- Department of Neuropathology and Prion Research, LMU Munich, Munich
| | | | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich
| | | | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg
| | | | | | | | | |
Collapse
|
23
|
Quach S, Holzgreve A, von Baumgarten L, Niyazi M, Unterrainer M, Thon N, Stöcklein S, Bartenstein P, Tonn JC, Albert NL. Increased TSPO PET signal after radiochemotherapy in IDH-wildtype glioma-indicator for treatment-induced immune activation? Eur J Nucl Med Mol Imaging 2022; 49:4282-4283. [PMID: 35610517 PMCID: PMC9525328 DOI: 10.1007/s00259-022-05844-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 01/23/2023]
Affiliation(s)
- Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany. .,Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK) partner site Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maximilian Niyazi
- German Cancer Consortium (DKTK) partner site Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Niklas Thon
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK) partner site Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sophia Stöcklein
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK) partner site Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK) partner site Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK) partner site Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
24
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
25
|
Havla J, Hohlfeld R. Antibody Therapies for Progressive Multiple Sclerosis and for Promoting Repair. Neurotherapeutics 2022; 19:774-784. [PMID: 35289375 PMCID: PMC9294105 DOI: 10.1007/s13311-022-01214-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/21/2022] Open
Abstract
Progressive multiple sclerosis (PMS) is clinically distinct from relapsing-remitting MS (RRMS). In PMS, clinical disability progression occurs independently of relapse activity. Furthermore, there is increasing evidence that the pathological mechanisms of PMS and RRMS are different. Current therapeutic options for the treatment of PMS remain inadequate, although ocrelizumab, a B-cell-depleting antibody, is now available as the first approved therapeutic option for primary progressive MS. Recent advances in understanding the pathophysiology of PMS provide hope for new innovative therapeutic options: these include antibody therapies with anti-inflammatory, neuroprotective, and/or remyelination-fostering effects. In this review, we summarize the relevant trial data relating to antibody therapy and consider future antibody options for treating PMS.
Collapse
Affiliation(s)
- Joachim Havla
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany.
- Data Integration for Future Medicine (DIFUTURE) Consortium, LMU Munich, Munich, Germany.
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
26
|
Vainio SK, Dickens AM, Matilainen M, López-Picón FR, Aarnio R, Eskola O, Solin O, Anthony DC, Rinne JO, Airas L, Haaparanta-Solin M. Dimethyl fumarate decreases short-term but not long-term inflammation in a focal EAE model of neuroinflammation. EJNMMI Res 2022; 12:6. [PMID: 35107664 PMCID: PMC8811048 DOI: 10.1186/s13550-022-00878-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
Background Dimethyl fumarate (DMF) is an oral immunomodulatory drug used in the treatment of autoimmune diseases. Here, we sought to study whether the effect of DMF can be detected using positron emission tomography (PET) targeting the 18-kDa translocator protein (TSPO) in the focal delayed-type hypersensitivity rat model of multiple sclerosis (fDTH-EAE). The rats were treated orally twice daily from lesion activation (day 0) with either vehicle (tap water with 0.08% Methocel, 200 µL; control group n = 4 (3 after week four)) or 15 mg/kg DMF (n = 4) in 0.08% aqueous Methocel (200 µL) for 8 weeks. The animals were imaged by PET using the TSPO tracer [18F]GE-180 in weeks 0, 1, 2, 4, 8, and 18 following lesion activation, and the non-displaceable binding potential (BPND) was calculated. Immunohistochemical staining for Iba1, CD4, and CD8 was performed in week 18, and in separate cohorts of animals, following 2 or 4 weeks of treatment. Results Using the fDTH-EAE model, DMF reduced the [18F]GE-180 BPND in the DMF-treated animals compared to control animals after 1 week of treatment (two-tailed unpaired t test, p = 0.031), but not in weeks 2, 4, 8, or 18 when imaged in vivo by PET. Immunostaining for Iba1 showed that DMF had no effect on the perilesional volume or the core lesion volume after 2 or 4 weeks of treatment, or at 18 weeks. However, the optical density (OD) measurements of CD4+ staining showed reduced OD in the lesions of the treated rats. Conclusions DMF reduced the microglial activation in the fDTH-EAE model after 1 week of treatment, as detected by PET imaging of the TSPO ligand [18F]GE-180. However, over an extended time course, reduced microglial activation was not observed using [18F]GE-180 or by immunohistochemistry for Iba1+ microglia/macrophages. Additionally, DMF did affect the infiltration of CD4+ and CD8+ T-lymphocytes at the fDTH-EAE lesion. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00878-y. In a focal rat DTH-EAE model of neuroinflammation, dimethyl fumarate decreases the uptake of TSPO PET tracer [18F]GE-180 in the short term. Long-term [18F]GE-180 follow-up did not indicate a treatment effect. Decreased neuroinflammation, CD4+ T cell infiltration, and CD8+ T cell infiltration were detected using immunohistochemistry.
Collapse
Affiliation(s)
- S K Vainio
- Turku PET Centre, Preclinical PET Imaging, Preclinical Imaging Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland. .,MediCity Research Laboratory, University of Turku, Turku, Finland.
| | - A M Dickens
- Department of Chemistry, University of Turku, Turku, Finland.,Turku Bioscience, Turku, Finland
| | - M Matilainen
- Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - F R López-Picón
- Turku PET Centre, Preclinical PET Imaging, Preclinical Imaging Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - R Aarnio
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - O Eskola
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - O Solin
- Accelerator Laboratory, Åbo Akademi University, Turku, Finland.,Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - D C Anthony
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - J O Rinne
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - L Airas
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland.,Department of Clinical Medicine, University of Turku, Turku, Finland
| | - M Haaparanta-Solin
- Turku PET Centre, Preclinical PET Imaging, Preclinical Imaging Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| |
Collapse
|
27
|
Li XG, Velikyan I, Viitanen R, Roivainen A. PET radiopharmaceuticals for imaging inflammatory diseases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00075-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
28
|
Kaiser L, Holzgreve A, Quach S, Ingrisch M, Unterrainer M, Dekorsy FJ, Lindner S, Ruf V, Brosch-Lenz J, Delker A, Böning G, Suchorska B, Niyazi M, Wetzel CH, Riemenschneider MJ, Stöcklein S, Brendel M, Rupprecht R, Thon N, von Baumgarten L, Tonn JC, Bartenstein P, Ziegler S, Albert NL. Differential Spatial Distribution of TSPO or Amino Acid PET Signal and MRI Contrast Enhancement in Gliomas. Cancers (Basel) 2021; 14:cancers14010053. [PMID: 35008218 PMCID: PMC8750092 DOI: 10.3390/cancers14010053] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 01/14/2023] Open
Abstract
Simple Summary Radiotracers targeting the translocator protein (TSPO) have recently gained substantial interest, since TSPO is overexpressed in malignant gliomas, where it correlates inversely with patient’s survival. The high-affinity TSPO PET ligand [18F]GE180 was found to depict tumor areas with a remarkably high contrast and has been shown to provide non-invasive information on histological tumor grades. Yet, its significance was questioned with the argument, that the high contrast may solely arise from nonspecific accumulation in tissue supplied by leaky vessels. This study aimed to address this question by providing a detailed evaluation of spatial associations between TSPO and amino acid PET with relative contrast enhancement in T1-weighted MRI. The results show that [18F]GE180 contrast does not reflect a disrupted blood–brain barrier (BBB) only and that multi-modal imaging generates complementary information, which may better depict spatial heterogeneity of tumor biology and may be used to individualize the therapy for each patient. Abstract In this study, dual PET and contrast enhanced MRI were combined to investigate their correlation per voxel in patients at initial diagnosis with suspected glioblastoma. Correlation with contrast enhancement (CE) as an indicator of BBB leakage was further used to evaluate whether PET signal is likely caused by BBB disruption alone, or rather attributable to specific binding after BBB passage. PET images with [18F]GE180 and the amino acid [18F]FET were acquired and normalized to healthy background (tumor-to-background ratio, TBR). Contrast enhanced images were normalized voxel by voxel with the pre-contrast T1-weighted MRI to generate relative CE values (rCE). Voxel-wise analysis revealed a high PET signal even within the sub-volumes without detectable CE. No to moderate correlation of rCE with TBR voxel-values and a small overlap as well as a larger distance of the hotspots delineated in rCE and TBR-PET images were detected. In contrast, voxel-wise correlation between both PET modalities was strong for most patients and hotspots showed a moderate overlap and distance. The high PET signal in tumor sub-volumes without CE observed in voxel-wise analysis as well as the discordant hotspots emphasize the specificity of the PET signals and the relevance of combined differential information from dual PET and MRI images.
Collapse
Affiliation(s)
- Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
- Correspondence:
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany; (S.Q.); (N.T.); (L.v.B.); (J.-C.T.)
| | - Michael Ingrisch
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.I.); (S.S.)
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.I.); (S.S.)
| | - Franziska J. Dekorsy
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | - Viktoria Ruf
- Center for Neuropathology and Prion Research, LMU Munich, 81377 Munich, Germany; (V.R.); (R.R.)
| | - Julia Brosch-Lenz
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | - Astrid Delker
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | - Guido Böning
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | | | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Christian H. Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany;
| | | | - Sophia Stöcklein
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.I.); (S.S.)
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany;
| | - Niklas Thon
- Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany; (S.Q.); (N.T.); (L.v.B.); (J.-C.T.)
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany; (S.Q.); (N.T.); (L.v.B.); (J.-C.T.)
| | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany; (S.Q.); (N.T.); (L.v.B.); (J.-C.T.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (A.H.); (M.U.); (F.J.D.); (S.L.); (J.B.-L.); (A.D.); (G.B.); (M.B.); (P.B.); (S.Z.); (N.L.A.)
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
29
|
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
|
30
|
Gelibter S, Pisa M, Croese T, Finardi A, Mandelli A, Sangalli F, Colombo B, Martinelli V, Comi G, Filippi M, Furlan R. Spinal Fluid Myeloid Microvesicles Predict Disease Course in Multiple Sclerosis. Ann Neurol 2021; 90:253-265. [PMID: 34216397 DOI: 10.1002/ana.26154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVE In vivo measures of myeloid activity are promising biomarkers in multiple sclerosis. We previously demonstrated that cerebrospinal fluid (CSF) myeloid microvesicles are markers of microglial/macrophage activity and neuroinflammation in multiple sclerosis. Here, we aimed at investigating the diagnostic and prognostic value of myeloid microvesicles in a clinical setting. METHODS Six hundred one patients discharged with a diagnosis of neuroinflammatory, neurodegenerative, or no neurological disease were enrolled. Myeloid microvesicles were measured with flow cytometry as isolectin B4-positive events in fresh CSF. Clinical, demographical, and magnetic resonance imaging (MRI) data were collected at diagnosis (all patients) and during follow-up (n = 176). RESULTS CSF myeloid microvesicles were elevated in neuroinflammatory patients compared to the neurodegenerative and control groups. In multiple sclerosis, microvesicles were higher in patients with MRI disease activity and their concentration increased along with the number of enhancing lesions (p < 0.0001, Jonckheere-Terpstra test). CSF myeloid microvesicles were also higher in patients with higher disease activity in the month and year preceding diagnosis. Microvesicles excellently discriminated between the relapsing-remitting and control groups (receiver operator characteristic curve, area under the curve = 0.939, p < 0.0001) and between radiologically isolated syndrome and unspecific brain lesions (0.942, p < 0.0001). Furthermore, microvesicles were independent predictors of prognosis for both the relapsing-remitting and progressive groups. Microvesicles independently predicted future disease activity in relapsing-remitting patients (hazard ratio [HR] = 1.967, 95% confidence interval [CI] = 1.147-3.372), correcting for prognostic factors of standard clinical use. In the progressive group, microvesicles were independent predictors of disability accrual (HR = 10.767, 95% CI = 1.335-86.812). INTERPRETATION Our results confirm that CSF myeloid microvesicles are a clinically meaningful biomarker of neuroinflammation and microglial/macrophage activity in vivo. These findings may support a possible use in clinical practice during diagnostic workup and prognostic assessment. ANN NEUROL 2021;90:253-265.
Collapse
Affiliation(s)
- Stefano Gelibter
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Marco Pisa
- Vita-Salute San Raffaele University, Milan, Italy
| | - Tommaso Croese
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Mandelli
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Bruno Colombo
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Massimo Filippi
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurorehabilitation Unit, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
31
|
Nutma E, Gebro E, Marzin MC, van der Valk P, Matthews PM, Owen DR, Amor S. Activated microglia do not increase 18 kDa translocator protein (TSPO) expression in the multiple sclerosis brain. Glia 2021; 69:2447-2458. [PMID: 34145928 PMCID: PMC8453709 DOI: 10.1002/glia.24052] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/20/2022]
Abstract
To monitor innate immune responses in the CNS, the 18 kDa Translocator protein (TSPO) is a frequently used target for PET imaging. The frequent assumption that increased TSPO expression in the human CNS reflects pro-inflammatory activation of microglia has been extrapolated from rodent studies. However, TSPO expression does not increase in activated human microglia in vitro. Studies of multiple sclerosis (MS) lesions reveal that TSPO is not restricted to pro-inflammatory microglia/macrophages, but also present in homeostatic or reparative microglia. Here, we investigated quantitative relationships between TSPO expression and microglia/macrophage phenotypes in white matter and lesions of brains with MS pathology. In white matter from brains with no disease pathology, normal appearing white matter (NAWM), active MS lesions and chronic active lesion rims, over 95% of TSPO+ cells are microglia/macrophages. Homeostatic microglial markers in NAWM and control tissue are lost/reduced in active lesions and chronic active lesion rims, reflecting cell activation. Nevertheless, pixel analysis of TSPO+ cells (n = 12,225) revealed that TSPO expression per cell is no higher in active lesions and chronic active lesion rims (where myeloid cells are activated) relative to NAWM and control. This data suggests that whilst almost all the TSPO signal in active lesions, chronic active lesion rims, NAWM and control is associated with microglia/macrophages, their TSPO expression predominantly reflects cell density and not activation phenotype. This finding has implications for the interpretation of TSPO PET signal in MS and other CNS diseases, and further demonstrates the limitation of extrapolating TSPO biology from rodents to humans.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, HV, Netherlands
| | - Emeline Gebro
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, HV, Netherlands
| | - Manuel C Marzin
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, HV, Netherlands
| | - Paul van der Valk
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, HV, Netherlands
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK.,UK Dementia Research Institute, Imperial College London, London, UK
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, HV, Netherlands.,Department of Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
32
|
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
|
33
|
Kimm MA, Klenk C, Alunni-Fabbroni M, Kästle S, Stechele M, Ricke J, Eisenblätter M, Wildgruber M. Tumor-Associated Macrophages-Implications for Molecular Oncology and Imaging. Biomedicines 2021; 9:biomedicines9040374. [PMID: 33918295 PMCID: PMC8066018 DOI: 10.3390/biomedicines9040374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) represent the largest group of leukocytes within the tumor microenvironment (TME) of solid tumors and orchestrate the composition of anti- as well as pro-tumorigenic factors. This makes TAMs an excellent target for novel cancer therapies. The plasticity of TAMs resulting in varying membrane receptors and expression of intracellular proteins allow the specific characterization of different subsets of TAMs. Those markers similarly allow tracking of TAMs by different means of molecular imaging. This review aims to provides an overview of the origin of tumor-associated macrophages, their polarization in different subtypes, and how characteristic markers of the subtypes can be used as targets for molecular imaging and theranostic approaches.
Collapse
Affiliation(s)
- Melanie A. Kimm
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Christopher Klenk
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Marianna Alunni-Fabbroni
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Sophia Kästle
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Matthias Stechele
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Michel Eisenblätter
- Department of Diagnostic and Interventional Radiology, Freiburg University Hospital, 79106 Freiburg, Germany;
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
- Correspondence: ; Tel.: +49-0-89-4400-76640
| |
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: 8] [Impact Index Per Article: 2.0] [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
|
Bagnato F, Gauthier SA, Laule C, Moore GRW, Bove R, Cai Z, Cohen-Adad J, Harrison DM, Klawiter EC, Morrow SA, Öz G, Rooney WD, Smith SA, Calabresi PA, Henry RG, Oh J, Ontaneda D, Pelletier D, Reich DS, Shinohara RT, Sicotte NL. Imaging Mechanisms of Disease Progression in Multiple Sclerosis: Beyond Brain Atrophy. J Neuroimaging 2021; 30:251-266. [PMID: 32418324 DOI: 10.1111/jon.12700] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
Clinicians involved with different aspects of the care of persons with multiple sclerosis (MS) and scientists with expertise on clinical and imaging techniques convened in Dallas, TX, USA on February 27, 2019 at a North American Imaging in Multiple Sclerosis Cooperative workshop meeting. The aim of the workshop was to discuss cardinal pathobiological mechanisms implicated in the progression of MS and novel imaging techniques, beyond brain atrophy, to unravel these pathologies. Indeed, although brain volume assessment demonstrates changes linked to disease progression, identifying the biological mechanisms leading up to that volume loss are key for understanding disease mechanisms. To this end, the workshop focused on the application of advanced magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging techniques to assess and measure disease progression in both the brain and the spinal cord. Clinical translation of quantitative MRI was recognized as of vital importance, although the need to maintain a relatively short acquisition time mandated by most radiology departments remains the major obstacle toward this effort. Regarding PET, the panel agreed upon its utility to identify ongoing pathological processes. However, due to costs, required expertise, and the use of ionizing radiation, PET was not considered to be a viable option for ongoing care of persons with MS. Collaborative efforts fostering robust study designs and imaging technique standardization across scanners and centers are needed to unravel disease mechanisms leading to progression and discovering medications halting neurodegeneration and/or promoting repair.
Collapse
Affiliation(s)
- Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Susan A Gauthier
- Judith Jaffe Multiple Sclerosis Center, Department of Neurology, Feil Family Brain and Mind Institute, and Department of Radiology, Weill Cornell Medicine, New York, NY
| | - Cornelia Laule
- Department of Radiology, Pathology, and Laboratory Medicine, Department of Physics and Astronomy, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - George R Wayne Moore
- Department of Pathology and Laboratory Medicine, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Riley Bove
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University, New Haven, CT
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal and Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Quebec, Canada
| | - Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD
| | - Eric C Klawiter
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sarah A Morrow
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Gülin Öz
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN
| | - William D Rooney
- Advanced Imaging Research Center, Departments of Biomedical Engineering, Neurology, and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | - Seth A Smith
- Radiology and Radiological Sciences and Vanderbilt University Imaging Institute, Vanderbilt University Medical Center, and Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Roland G Henry
- Departments of Neurology, Radiology and Biomedical Imaging, and the UC San Francisco & Berkeley Bioengineering Graduate Group, University of California San Francisco, San Francisco, CA
| | - Jiwon Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Neurology, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Daniel Pelletier
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
| | -
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
37
|
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: 103] [Impact Index Per Article: 25.8] [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
|
38
|
TSPO PET With 18F-GE-180 to Differentiate Variants of Multiple Sclerosis: Relapsing-Remitting Multiple Sclerosis, Tumefactive Demyelination, and Baló's Concentric Sclerosis. Clin Nucl Med 2020; 45:e447-e448. [PMID: 32796248 DOI: 10.1097/rlu.0000000000003220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PET targeting the translocator protein (TSPO) expression is an interesting approach to detect neuroinflammation, as TSPO is upregulated in activated macrophages and microglia. Considering the variable pathophysiology of multiple sclerosis (MS) variants, we compare TSPO PET using F-GE-180 in 3 different demyelinating diseases of the central nervous system: relapsing-remitting MS, tumefactive MS, and Baló's concentric sclerosis. Visualization of neuroinflammation and its PET patterns in addition to MRI may contribute to accurate distinction and monitoring of central nervous system demyelination.
Collapse
|
39
|
Nicholas R, Brooks D, Owen D. 18F-GE180, a radioligand for the TSPO protein: not ready for clinical trials in multiple sclerosis. Eur J Nucl Med Mol Imaging 2020; 47:2242-2243. [PMID: 32383091 DOI: 10.1007/s00259-020-04844-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/27/2020] [Indexed: 01/24/2023]
Affiliation(s)
- Richard Nicholas
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK. .,Imperial College London, Charing Cross Hospital, Room 10L18, Fulham Palace Road, London, W6 8RF, UK.
| | - David Brooks
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David Owen
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
40
|
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
|
41
|
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
|
42
|
Chen BY, Ghezzi C, Villegas B, Quon A, Radu CG, Witte ON, Clark PM. 18F-FAC PET Visualizes Brain-Infiltrating Leukocytes in a Mouse Model of Multiple Sclerosis. J Nucl Med 2020; 61:757-763. [PMID: 31653711 PMCID: PMC7198381 DOI: 10.2967/jnumed.119.229351] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 10/07/2019] [Indexed: 12/26/2022] Open
Abstract
Brain-infiltrating leukocytes contribute to multiple sclerosis (MS) and autoimmune encephalomyelitis and likely play a role in traumatic brain injury, seizure, and stroke. Brain-infiltrating leukocytes are also primary targets for MS disease-modifying therapies. However, no method exists for noninvasively visualizing these cells in a living organism. 1-(2'-deoxy-2'-18F-fluoroarabinofuranosyl) cytosine (18F-FAC) is a PET radiotracer that measures deoxyribonucleoside salvage and accumulates preferentially in immune cells. We hypothesized that 18F-FAC PET could noninvasively image brain-infiltrating leukocytes. Methods: Healthy mice were imaged with 18F-FAC PET to quantify if this radiotracer crosses the blood-brain barrier (BBB). Experimental autoimmune encephalomyelitis (EAE) is a mouse disease model with brain-infiltrating leukocytes. To determine whether 18F-FAC accumulates in brain-infiltrating leukocytes, EAE mice were analyzed with 18F-FAC PET, digital autoradiography, and immunohistochemistry, and deoxyribonucleoside salvage activity in brain-infiltrating leukocytes was analyzed ex vivo. Fingolimod-treated EAE mice were imaged with 18F-FAC PET to assess if this approach can monitor the effect of an immunomodulatory drug on brain-infiltrating leukocytes. PET scans of individuals injected with 2-chloro-2'-deoxy-2'-18F-fluoro-9-β-d-arabinofuranosyl-adenine (18F-CFA), a PET radiotracer that measures deoxyribonucleoside salvage in humans, were analyzed to evaluate whether 18F-CFA crosses the human BBB. Results:18F-FAC accumulates in the healthy mouse brain at levels similar to 18F-FAC in the blood (2.54 ± 0.2 and 3.04 ± 0.3 percentage injected dose per gram, respectively) indicating that 18F-FAC crosses the BBB. EAE mice accumulate 18F-FAC in the brain at 180% of the levels of control mice. Brain 18F-FAC accumulation localizes to periventricular regions with significant leukocyte infiltration, and deoxyribonucleoside salvage activity is present at similar levels in brain-infiltrating T and innate immune cells. These data suggest that 18F-FAC accumulates in brain-infiltrating leukocytes in this model. Fingolimod-treated EAE mice accumulate 18F-FAC in the brain at 37% lower levels than control-treated EAE mice, demonstrating that 18F-FAC PET can monitor therapeutic interventions in this mouse model. 18F-CFA accumulates in the human brain at 15% of blood levels (0.08 ± 0.01 and 0.54 ± 0.07 SUV, respectively), indicating that 18F-CFA does not cross the BBB in humans. Conclusion:18F-FAC PET can visualize brain-infiltrating leukocytes in a mouse MS model and can monitor the response of these cells to an immunomodulatory drug. Translating this strategy into humans will require exploring additional radiotracers.
Collapse
Affiliation(s)
- Bao Ying Chen
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California
| | - Chiara Ghezzi
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California
| | - Brendon Villegas
- Department of Pulmonary and Critical Care Medicine, UCLA, Los Angeles, California
| | - Andrew Quon
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Ahmanson Translational Imaging Division, UCLA, Los Angeles, California
| | - Caius G Radu
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Ahmanson Translational Imaging Division, UCLA, Los Angeles, California
| | - Owen N Witte
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California; and
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California
| | - Peter M Clark
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California; and
| |
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
|
Cavaliere C, Tramontano L, Fiorenza D, Alfano V, Aiello M, Salvatore M. Gliosis and Neurodegenerative Diseases: The Role of PET and MR Imaging. Front Cell Neurosci 2020; 14:75. [PMID: 32327973 PMCID: PMC7161920 DOI: 10.3389/fncel.2020.00075] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/13/2020] [Indexed: 12/16/2022] Open
Abstract
Glial activation characterizes most neurodegenerative and psychiatric diseases, often anticipating clinical manifestations and macroscopical brain alterations. Although imaging techniques have improved diagnostic accuracy in many neurological conditions, often supporting diagnosis, prognosis prediction and treatment outcome, very few molecular imaging probes, specifically focused on microglial and astrocytic activation, have been translated to a clinical setting. In this context, hybrid positron emission tomography (PET)/magnetic resonance (MR) scanners represent the most advanced tool for molecular imaging, combining the functional specificity of PET radiotracers (e.g., targeting metabolism, hypoxia, and inflammation) to both high-resolution and multiparametric information derived by MR in a single imaging acquisition session. This simultaneity of findings achievable by PET/MR, if useful for reciprocal technical adjustments regarding temporal and spatial cross-modal alignment/synchronization, opens still debated issues about its clinical value in neurological patients, possibly incompliant and highly variable from a clinical point of view. While several preclinical and clinical studies have investigated the sensitivity of PET tracers to track microglial (mainly TSPO ligands) and astrocytic (mainly MAOB ligands) activation, less studies have focused on MR specificity to this topic (e.g., through the assessment of diffusion properties and T2 relaxometry), and only few exploiting the integration of simultaneous hybrid acquisition. This review aims at summarizing and critically review the current state about PET and MR imaging for glial targets, as well as the potential added value of hybrid scanners for characterizing microglial and astrocytic activation.
Collapse
|
46
|
Werner JM, Lohmann P, Fink GR, Langen KJ, Galldiks N. Current Landscape and Emerging Fields of PET Imaging in Patients with Brain Tumors. Molecules 2020; 25:E1471. [PMID: 32213992 PMCID: PMC7146177 DOI: 10.3390/molecules25061471] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The number of positron-emission tomography (PET) tracers used to evaluate patients with brain tumors has increased substantially over the last years. For the management of patients with brain tumors, the most important indications are the delineation of tumor extent (e.g., for planning of resection or radiotherapy), the assessment of treatment response to systemic treatment options such as alkylating chemotherapy, and the differentiation of treatment-related changes (e.g., pseudoprogression or radiation necrosis) from tumor progression. Furthermore, newer PET imaging approaches aim to address the need for noninvasive assessment of tumoral immune cell infiltration and response to immunotherapies (e.g., T-cell imaging). This review summarizes the clinical value of the landscape of tracers that have been used in recent years for the above-mentioned indications and also provides an overview of promising newer tracers for this group of patients.
Collapse
Affiliation(s)
- Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Gereon R. Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
- Department of Nuclear Medicine, University Hospital Aachen, 52074 Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| |
Collapse
|
47
|
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]
|
48
|
Unterrainer M, Fleischmann DF, Vettermann F, Ruf V, Kaiser L, Nelwan D, Lindner S, Brendel M, Wenter V, Stöcklein S, Herms J, Milenkovic VM, Rupprecht R, Tonn JC, Belka C, Bartenstein P, Niyazi M, Albert NL. TSPO PET, tumour grading and molecular genetics in histologically verified glioma: a correlative 18F-GE-180 PET study. Eur J Nucl Med Mol Imaging 2019; 47:1368-1380. [PMID: 31486876 DOI: 10.1007/s00259-019-04491-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND The 18-kDa translocator protein (TSPO) is overexpressed in brain tumours and represents an interesting target for glioma imaging. 18F-GE-180, a novel TSPO ligand, has shown improved binding affinity and a high target-to-background contrast in patients with glioblastoma. However, the association of uptake characteristics on TSPO PET using 18F-GE-180 with the histological WHO grade and molecular genetic features so far remains unknown and was evaluated in the current study. METHODS Fifty-eight patients with histologically validated glioma at initial diagnosis or recurrence were included. All patients underwent 18F-GE-180 PET, and the maximal and mean tumour-to-background ratios (TBRmax, TBRmean) as well as the PET volume were assessed. On MRI, presence/absence of contrast enhancement was evaluated. Imaging characteristics were correlated with neuropathological parameters (i.e. WHO grade, isocitrate dehydrogenase (IDH) mutation, O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation and telomerase reverse transcriptase (TERT) promoter mutation). RESULTS Six of 58 patients presented with WHO grade II, 16/58 grade III and 36/58 grade IV gliomas. An (IDH) mutation was found in 19/58 cases, and 39/58 were classified as IDH-wild type. High 18F-GE-180-uptake was observed in all but 4 cases (being WHO grade II glioma, IDH-mutant). A high association of 18F-GE-180-uptake and WHO grades was seen: WHO grade IV gliomas showed the highest uptake intensity compared with grades III and II gliomas (median TBRmax 5.15 (2.59-8.95) vs. 3.63 (1.85-7.64) vs. 1.63 (1.50-3.43), p < 0.001); this association with WHO grades persisted within the IDH-wild-type and IDH-mutant subgroup analyses (p < 0.05). Uptake intensity was also associated with the IDH mutational status with a trend towards higher 18F-GE-180-uptake in IDH-wild-type gliomas in the overall group (median TBRmax 4.67 (1.56-8.95) vs. 3.60 (1.50-7.64), p = 0.083); however, within each WHO grade, no differences were found (e.g. median TBRmax in WHO grade III glioma 4.05 (1.85-5.39) vs. 3.36 (2.32-7.64), p = 1.000). No association was found between uptake intensity and MGMT or TERT (p > 0.05 each). CONCLUSION Uptake characteristics on 18F-GE-180 PET are highly associated with the histological WHO grades, with the highest 18F-GE-180 uptake in WHO grade IV glioblastomas and a PET-positive rate of 100% among the investigated high-grade gliomas. Conversely, all TSPO-negative cases were WHO grade II gliomas. The observed association of 18F-GE-180 uptake and the IDH mutational status seems to be related to the high inter-correlation of the IDH mutational status and the WHO grades.
Collapse
Affiliation(s)
- M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D F Fleischmann
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - F Vettermann
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - V Ruf
- Department of Neuropathology, LMU Munich, Munich, Germany
| | - L Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - D Nelwan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - S Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - M Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - V Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - S Stöcklein
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - J Herms
- Department of Neuropathology, LMU Munich, Munich, Germany
| | - V M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - R Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - J C Tonn
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - C Belka
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Niyazi
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - N L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
49
|
Chronic inflammation in multiple sclerosis - seeing what was always there. Nat Rev Neurol 2019; 15:582-593. [PMID: 31420598 DOI: 10.1038/s41582-019-0240-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 12/18/2022]
Abstract
Activation of innate immune cells and other compartmentalized inflammatory cells in the brains and spinal cords of people with relapsing-remitting multiple sclerosis (MS) and progressive MS has been well described histopathologically. However, conventional clinical MRI is largely insensitive to this inflammatory activity. The past two decades have seen the introduction of quantitative dynamic MRI scanning with contrast agents that are sensitive to the reduction in blood-brain barrier integrity associated with inflammation and to the trafficking of inflammatory myeloid cells. New MRI imaging sequences provide improved contrast for better detection of grey matter lesions. Quantitative lesion volume measures and magnetic resonance susceptibility imaging are sensitive to the activity of macrophages in the rims of white matter lesions. PET and magnetic resonance spectroscopy methods can also be used to detect contributions from innate immune activation in the brain and spinal cord. Some of these advanced research imaging methods for visualization of chronic inflammation are practical for relatively routine clinical applications. Observations made with the use of these techniques suggest ways of stratifying patients with MS to improve their care. The imaging methods also provide new tools to support the development of therapies for chronic inflammation in MS.
Collapse
|
50
|
Recent Developments in TSPO PET Imaging as A Biomarker of Neuroinflammation in Neurodegenerative Disorders. Int J Mol Sci 2019; 20:ijms20133161. [PMID: 31261683 PMCID: PMC6650818 DOI: 10.3390/ijms20133161] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is an inflammatory response in the brain and spinal cord, which can involve the activation of microglia and astrocytes. It is a common feature of many central nervous system disorders, including a range of neurodegenerative disorders. An overlap between activated microglia, pro-inflammatory cytokines and translocator protein (TSPO) ligand binding was shown in early animal studies of neurodegeneration. These findings have been translated in clinical studies, where increases in TSPO positron emission tomography (PET) signal occur in disease-relevant areas across a broad spectrum of neurodegenerative diseases. While this supports the use of TSPO PET as a biomarker to monitor response in clinical trials of novel neurodegenerative therapeutics, the clinical utility of current TSPO PET radioligands has been hampered by the lack of high affinity binding to a prevalent form of polymorphic TSPO (A147T) compared to wild type TSPO. This review details recent developments in exploration of ligand-sensitivity to A147T TSPO that have yielded ligands with improved clinical utility. In addition to developing a non-discriminating TSPO ligand, the final frontier of TSPO biomarker research requires developing an understanding of the cellular and functional interpretation of the TSPO PET signal. Recent insights resulting from single cell analysis of microglial phenotypes are reviewed.
Collapse
|