1
|
Bisi N, Pinzi L, Rastelli G, Tonali N. Early Diagnosis of Neurodegenerative Diseases: What Has Been Undertaken to Promote the Transition from PET to Fluorescence Tracers. Molecules 2024; 29:722. [PMID: 38338465 PMCID: PMC10856728 DOI: 10.3390/molecules29030722] [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: 01/04/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Alzheimer's Disease (AD) and Parkinson's Disease (PD) represent two among the most frequent neurodegenerative diseases worldwide. A common hallmark of these pathologies is the misfolding and consequent aggregation of amyloid proteins into soluble oligomers and insoluble β-sheet-rich fibrils, which ultimately lead to neurotoxicity and cell death. After a hundred years of research on the subject, this is the only reliable histopathological feature in our hands. Since AD and PD are diagnosed only once neuronal death and the first symptoms have appeared, the early detection of these diseases is currently impossible. At present, there is no effective drug available, and patients are left with symptomatic and inconclusive therapies. Several reasons could be associated with the lack of effective therapeutic treatments. One of the most important factors is the lack of selective probes capable of detecting, as early as possible, the most toxic amyloid species involved in the onset of these pathologies. In this regard, chemical probes able to detect and distinguish among different amyloid aggregates are urgently needed. In this article, we will review and put into perspective results from ex vivo and in vivo studies performed on compounds specifically interacting with such early species. Following a general overview on the three different amyloid proteins leading to insoluble β-sheet-rich amyloid deposits (amyloid β1-42 peptide, Tau, and α-synuclein), a list of the advantages and disadvantages of the approaches employed to date is discussed, with particular attention paid to the translation of fluorescence imaging into clinical applications. Furthermore, we also discuss how the progress achieved in detecting the amyloids of one neurodegenerative disease could be leveraged for research into another amyloidosis. As evidenced by a critical analysis of the state of the art, substantial work still needs to be conducted. Indeed, the early diagnosis of neurodegenerative diseases is a priority, and we believe that this review could be a useful tool for better investigating this field.
Collapse
Affiliation(s)
- Nicolò Bisi
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Nicolò Tonali
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| |
Collapse
|
2
|
Pijeira MSO, Nunes PSG, Chaviano SL, Diaz AMA, DaSilva JN, Ricci-Junior E, Alencar LMR, Chen X, Santos-Oliveira R. Medicinal (Radio) Chemistry: Building Radiopharmaceuticals for the Future. Curr Med Chem 2024; 31:5481-5534. [PMID: 37594105 DOI: 10.2174/0929867331666230818092634] [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: 03/10/2023] [Revised: 05/30/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Radiopharmaceuticals are increasingly playing a leading role in diagnosing, monitoring, and treating disease. In comparison with conventional pharmaceuticals, the development of radiopharmaceuticals does follow the principles of medicinal chemistry in the context of imaging-altered physiological processes. The design of a novel radiopharmaceutical has several steps similar to conventional drug discovery and some particularity. In the present work, we revisited the insights of medicinal chemistry in the current radiopharmaceutical development giving examples in oncology, neurology, and cardiology. In this regard, we overviewed the literature on radiopharmaceutical development to study overexpressed targets such as prostate-specific membrane antigen and fibroblast activation protein in cancer; β-amyloid plaques and tau protein in brain disorders; and angiotensin II type 1 receptor in cardiac disease. The work addresses concepts in the field of radiopharmacy with a special focus on the potential use of radiopharmaceuticals for nuclear imaging and theranostics.
Collapse
Affiliation(s)
- Martha Sahylí Ortega Pijeira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
| | - Paulo Sérgio Gonçalves Nunes
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas SP13083-970, Brazil
| | - Samila Leon Chaviano
- Laboratoire de Biomatériaux pour l'Imagerie Médicale, Axe Médicine Régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Aida M Abreu Diaz
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean N DaSilva
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Eduardo Ricci-Junior
- Laboratório de Desenvolvimento Galênico, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Laboratory of Biophysics and Nanosystems, Federal University of Maranhão, Av. dos Portugueses, 1966, Vila Bacanga, São Luís MA65080-805, Brazil
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore 117597, Singapore
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro State University, Rio de Janeiro 23070200, Brazil
| |
Collapse
|
3
|
Zhang S, Dong H, Bian J, Li D, Liu C. Targeting amyloid proteins for clinical diagnosis of neurodegenerative diseases. FUNDAMENTAL RESEARCH 2023; 3:505-519. [PMID: 38933553 PMCID: PMC11197785 DOI: 10.1016/j.fmre.2022.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Abnormal aggregation and accumulation of pathological amyloid proteins such as amyloid-β, Tau, and α-synuclein play key pathological roles and serve as histological hallmarks in different neurodegenerative diseases (NDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, various post-translational modifications (PTMs) have been identified on pathological amyloid proteins and are subjected to change during disease progression. Given the central role of amyloid proteins in NDs, tremendous efforts have been made to develop amyloid-targeting strategies for clinical diagnosis and molecular classification of NDs. In this review, we summarize two major strategies for targeting amyloid aggregates, with a focus on the trials in AD diagnosis. The first strategy is a positron emission tomography (PET) scan of protein aggregation in the brain. We mainly focus on introducing the development of small-molecule PET tracers for specifically recognizing pathological amyloid fibrils. The second strategy is the detection of PTM biomarkers on amyloid proteins in cerebrospinal fluid and plasma. We discuss the pathological roles of different PTMs in diseases and how we can use the PTM profile of amyloid proteins for clinical diagnosis. Finally, we point out the potential technical challenges of these two strategies, and outline other potential strategies, as well as a combination of multiple strategies, for molecular diagnosis of NDs.
Collapse
Affiliation(s)
- Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hui Dong
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Bian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
4
|
Dang C, Wang Y, Li Q, Lu Y. Neuroimaging modalities in the detection of Alzheimer's disease-associated biomarkers. PSYCHORADIOLOGY 2023; 3:kkad009. [PMID: 38666112 PMCID: PMC11003434 DOI: 10.1093/psyrad/kkad009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/04/2023] [Accepted: 06/20/2023] [Indexed: 04/28/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Neuropathological changes in AD patients occur up to 10-20 years before the emergence of clinical symptoms. Specific diagnosis and appropriate intervention strategies are crucial during the phase of mild cognitive impairment (MCI) and AD. The detection of biomarkers has emerged as a promising tool for tracking the efficacy of potential therapies, making an early disease diagnosis, and prejudging treatment prognosis. Specifically, multiple neuroimaging modalities, including magnetic resonance imaging (MRI), positron emission tomography, optical imaging, and single photon emission-computed tomography, have provided a few potential biomarkers for clinical application. The MRI modalities described in this review include structural MRI, functional MRI, diffusion tensor imaging, magnetic resonance spectroscopy, and arterial spin labelling. These techniques allow the detection of presymptomatic diagnostic biomarkers in the brains of cognitively normal elderly people and might also be used to monitor AD disease progression after the onset of clinical symptoms. This review highlights potential biomarkers, merits, and demerits of different neuroimaging modalities and their clinical value in MCI and AD patients. Further studies are necessary to explore more biomarkers and overcome the limitations of multiple neuroimaging modalities for inclusion in diagnostic criteria for AD.
Collapse
Affiliation(s)
- Chun Dang
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanchao Wang
- Department of Neurology, Chifeng University of Affiliated Hospital, Chifeng 024000, China
| | - Qian Li
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Yaoheng Lu
- Department of General Surgery, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Chengdu 610000, China
| |
Collapse
|
5
|
Lin HC, Lin KJ, Huang KL, Chen SH, Ho TY, Huang CC, Hsu JL, Chang CC, Hsiao IT. Visual reading for [ 18F]Florzolotau ([ 18F]APN-1607) tau PET imaging in clinical assessment of Alzheimer's disease. Front Neurosci 2023; 17:1148054. [PMID: 37250400 PMCID: PMC10213356 DOI: 10.3389/fnins.2023.1148054] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Tau-targeted positron emission tomography (tau-PET) is a potential tool for the differential diagnosis of Alzheimer's disease (AD) and to clarify the distribution of tau deposition. In addition to the quantitative analysis of tau-PET scans, visual reading supports the assessment of tau loading for clinical diagnosis. This study aimed to propose a method for visually interpreting tau-PET using the [18F] Florzolotau tracer and investigate the performance and utility of the visual reading. Materials and methods A total number of 46 individuals with 12 cognitively unimpaired subjects (CU), 20 AD patients with mild cognitive impairment (AD-MCI), and 14 AD with dementia (AD-D) patients with both [18F]Florbetapir amyloid PET and [18F]Florzolotau tau PET scans were included. Clinical information, cognitive assessment, and amyloid PET scan results were recorded. For visual interpretation, a modified rainbow colormap was created and a regional tau uptake scoring system was proposed to evaluate the degree of tracer uptake and its spatial distribution within five cortical regions. Each region was scored on a scale of [0, 2] as compared to the background, and that resulted in a global scale range of [0, 10]. Four readers interpreted [18F]Florzolotau PET using the visual scale. The global and regional standardized uptake value ratios (SUVr) were also calculated for analysis. Results The result indicates the average global visual scores were 0 ± 0 in the CU group, 3.43 ± 3.35 in the AD-MCI group, and 6.31 ± 2.97 in the AD-D group (p < 0.001). The consensus among the four observers on image scores was high with an intraclass correlation coefficient of 0.880 (95% CI: 0.767-0.936). The average global visual score was significantly associated with global SUVr (r = 0.884, p < 0.0001) and with the CDR-sum of box (r = 0.677, p < 0.0001). Conclusion The visual reading method generated a visual score of [18F]Florzolotau tau-PET with good sensitivity and specificity to identify AD-D or CU individuals from the other patients. The preliminary result also showed that the global visual scores are significantly and reliably correlated with global cortical SUVr, and associated well with the clinical diagnosis and cognitive performance.
Collapse
Affiliation(s)
- Huan-Chun Lin
- Department of Nuclear Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| | - Kuo-Lun Huang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Shih-Hsin Chen
- Department of Nuclear Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| | - Tsung-Ying Ho
- Department of Nuclear Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| | - Chin-Chang Huang
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Jung-Lung Hsu
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Neurology, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei City, Taiwan
| | - Chiung-Chih Chang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ing-Tsung Hsiao
- Department of Nuclear Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| |
Collapse
|
6
|
Sarabia-Vallejo Á, López-Alvarado P, Menéndez JC. Small-molecule theranostics in Alzheimer's disease. Eur J Med Chem 2023; 255:115382. [PMID: 37141706 DOI: 10.1016/j.ejmech.2023.115382] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/06/2023]
Abstract
Alzheimer's Disease (AD) remains one of the most challenging health-related issues for our society. It is becoming increasingly prevalent, especially in developed countries, due to the rising life expectancy and, moreover, represents a considerable economic burden worldwide. All efforts at the discovery of new diagnostic and therapeutic tools in the last decades have invariably met with failure, making AD an incurable illness and underscoring the need for new approaches. In recent years, theranostic agents have emerged as an interesting strategy. They are molecules able to simultaneously provide diagnostic information and deliver therapeutic activity, allowing for the assessment of the molecule activity, the organism response and the pharmacokinetics. This makes these compounds promising for streamlining research on AD drugs and for their application in personalized medicine. We review here the field of small-molecule theranostic agents as promising tools for the development of novel diagnostic and therapeutic resources against AD, highlighting the positive and significant impact that theranostics can be expected to have in the near future in clinical practice.
Collapse
Affiliation(s)
- Álvaro Sarabia-Vallejo
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Pilar López-Alvarado
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - J Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain.
| |
Collapse
|
7
|
Mohammadi Z, Alizadeh H, Marton J, Cumming P. The Sensitivity of Tau Tracers for the Discrimination of Alzheimer's Disease Patients and Healthy Controls by PET. Biomolecules 2023; 13:290. [PMID: 36830659 PMCID: PMC9953528 DOI: 10.3390/biom13020290] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Hyperphosphorylated tau aggregates, also known as neurofibrillary tangles, are a hallmark neuropathological feature of Alzheimer's disease (AD). Molecular imaging of tau by positron emission tomography (PET) began with the development of [18F]FDDNP, an amyloid β tracer with off-target binding to tau, which obtained regional specificity through the differing distributions of amyloid β and tau in AD brains. A concerted search for more selective and affine tau PET tracers yielded compounds belonging to at least eight structural categories; 18F-flortaucipir, known variously as [18F]-T807, AV-1451, and Tauvid®, emerged as the first tau tracer approved by the American Food and Drug Administration. The various tau tracers differ concerning their selectivity over amyloid β, off-target binding at sites such as monoamine oxidase and neuromelanin, and degree of uptake in white matter. While there have been many reviews of molecular imaging of tau in AD and other conditions, there has been no systematic comparison of the fitness of the various tracers for discriminating between AD patient and healthy control (HC) groups. In this narrative review, we endeavored to compare the binding properties of the various tau tracers in vitro and the effect size (Cohen's d) for the contrast by PET between AD patients and age-matched HC groups. The available tracers all gave good discrimination, with Cohen's d generally in the range of two-three in culprit brain regions. Overall, Cohen's d was higher for AD patient groups with more severe illness. Second-generation tracers, while superior concerning off-target binding, do not have conspicuously higher sensitivity for the discrimination of AD and HC groups. We suppose that available pharmacophores may have converged on a maximal affinity for tau fibrils, which may limit the specific signal imparted in PET studies.
Collapse
Affiliation(s)
- Zohreh Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hadi Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Straße 10-14, D-01454 Radeberg, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
8
|
Hoenig MC, Drzezga A. Clear-headed into old age: Resilience and resistance against brain aging-A PET imaging perspective. J Neurochem 2023; 164:325-345. [PMID: 35226362 DOI: 10.1111/jnc.15598] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 11/28/2022]
Abstract
With the advances in modern medicine and the adaptation towards healthier lifestyles, the average life expectancy has doubled since the 1930s, with individuals born in the millennium years now carrying an estimated life expectancy of around 100 years. And even though many individuals around the globe manage to age successfully, the prevalence of aging-associated neurodegenerative diseases such as sporadic Alzheimer's disease has never been as high as nowadays. The prevalence of Alzheimer's disease is anticipated to triple by 2050, increasing the societal and economic burden tremendously. Despite all efforts, there is still no available treatment defeating the accelerated aging process as seen in this disease. Yet, given the advances in neuroimaging techniques that are discussed in the current Review article, such as in positron emission tomography (PET) or magnetic resonance imaging (MRI), pivotal insights into the heterogenous effects of aging-associated processes and the contribution of distinct lifestyle and risk factors already have and are still being gathered. In particular, the concepts of resilience (i.e. coping with brain pathology) and resistance (i.e. avoiding brain pathology) have more recently been discussed as they relate to mechanisms that are associated with the prolongation and/or even stop of the progressive brain aging process. Better understanding of the underlying mechanisms of resilience and resistance may one day, hopefully, support the identification of defeating mechanism against accelerating aging.
Collapse
Affiliation(s)
- Merle C Hoenig
- Research Center Juelich, Institute for Neuroscience and Medicine II, Molecular Organization of the Brain, Juelich, Germany.,Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Alexander Drzezga
- Research Center Juelich, Institute for Neuroscience and Medicine II, Molecular Organization of the Brain, Juelich, Germany.,Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases, Bonn/Cologne, Germany
| |
Collapse
|
9
|
Liu T, Li Y, Wang Y, Yan XX, Dai J, Cui M. Discovery and evaluation of aza-fused tricyclic derivatives for detection of Tau pathology in Alzheimer's disease. Eur J Med Chem 2023; 246:114991. [PMID: 36493618 DOI: 10.1016/j.ejmech.2022.114991] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/09/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
For various neurodegenerative diseases, including Alzheimer's disease (AD), the abnormal aggregation of Tau is not only the predominant contributing factor but also a major biomarker for disease diagnosis. In this study, a series of aza-fused tricyclic derivatives were designed and synthesized. By changing the position and number of nitrogen atoms on the fused tricyclic core, the imidazonaphthyridine scaffold was screened and reported for the first time which could potentially detect Tau aggregates. Through a series of in vitro and in vivo biological evaluations, probe [125I]5 possessed exceptional binding affinity (IC50 = 1.63 nM) to neurofibrillary tangles in the AD brain, high selectivity over Aβ plaques (23.4-fold), clean off-target profile to monoamine oxidase A/B (MAO-A/B), and suitable pharmacokinetics (initial brain uptake = 3.22% ID/g).
Collapse
Affiliation(s)
- Tianqing Liu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Yuying Li
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Yan Wang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, China
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan, 430074, China
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China; Center for Advanced Materials Research, Beijing Normal University, Zhuhai, 519087, China.
| |
Collapse
|
10
|
Pierre K, Molina V, Shukla S, Avila A, Fong N, Nguyen J, Lucke-Wold B. Chronic traumatic encephalopathy: Diagnostic updates and advances. AIMS Neurosci 2022; 9:519-535. [PMID: 36660076 PMCID: PMC9826753 DOI: 10.3934/neuroscience.2022030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a progressive neurodegenerative disease that occurs secondary to repetitive mild traumatic brain injury. Current clinical diagnosis relies on symptomatology and structural imaging findings which often vary widely among those with the disease. The gold standard of diagnosis is post-mortem pathological examination. In this review article, we provide a brief introduction to CTE, current diagnostic workup and the promising research on imaging and fluid biomarker diagnostic techniques. For imaging, we discuss quantitative structural analyses, DTI, fMRI, MRS, SWI and PET CT. For fluid biomarkers, we discuss p-tau, TREM2, CCL11, NfL and GFAP.
Collapse
Affiliation(s)
- Kevin Pierre
- University of Florida Department of Radiology, Gainesville 32603, Florida, USA
| | - Vanessa Molina
- Sam Houston State University of Osteopathic Medicine, Conroe 77304, Texas, USA
| | - Shil Shukla
- Sam Houston State University of Osteopathic Medicine, Conroe 77304, Texas, USA
| | - Anthony Avila
- Sam Houston State University of Osteopathic Medicine, Conroe 77304, Texas, USA
| | - Nicholas Fong
- Sam Houston State University of Osteopathic Medicine, Conroe 77304, Texas, USA
| | - Jessica Nguyen
- Sam Houston State University of Osteopathic Medicine, Conroe 77304, Texas, USA
| | - Brandon Lucke-Wold
- University of Florida Department of Neurosurgery, Gainesville 32603, Florida, USA,* Correspondence:
| |
Collapse
|
11
|
Fu L, Zhang J, Zhou K, Zhang X, Xie H, Zhu M, Cui M, Wang R. In vivo imaging of tau deposition in Alzheimer’s disease using both [18F]-THK5317 and [18F]-S16: A pilot human study. Front Aging Neurosci 2022; 14:994750. [PMID: 36092808 PMCID: PMC9459225 DOI: 10.3389/fnagi.2022.994750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To evaluate the effectiveness of a new tracer (S)-1-(4-(6-(dimethylamino)quinoxalin-2-yl)phenoxy)-3-fluoropropan-2-ol ([18F]-S16), in distinguishing patients with AD from HCs. Methods Paired [18F]-S16 and [18F]-THK5317 scans were acquired in five patients with AD, six HCs, one subject with a semantic variant of primary progressive aphasia (sv-PPA) and one subject with probable progressive supranuclear palsy (PSP). Dynamic PET scanning was performed over 90 min after injection of the tracers. Standardized uptake values (SUV) and cortical-to-cerebellum standardized uptake value ratios (SUVRs) were used for tau deposition semi-quantization. A voxel-based analysis was employed to assess the uptake difference between populations. Results [18F]-S16 exhibited excellent blood-brain-barrier penetration. AD patients showed increased cortical [18F]-THK5317 and [18F]-S16 binding. Compared to HCs, AD patients showed significantly increased cortical [18F]-S16 uptake in the bilateral occipital cortex, posterior cingulated cortex/precuneus, and lateral frontal cortex. Notable [18F]-S16 uptake was observed in the basal ganglia and brainstem compared to the neocortex. A substantial [18F]-S16 signal was detected in the basal ganglia and midbrain in a patient with probable PSP and in the bilateral anterior temporal cortex in a sv-PPA patient. Conclusion [18F]-S16 might be of help to detect tau protein in vivo.
Collapse
Affiliation(s)
- Liping Fu
- Department of Nuclear Medicine, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Liping Fu,
| | - Jinming Zhang
- Department of Nuclear Medicine, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Kaixiang Zhou
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hengge Xie
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mingwei Zhu
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Ruimin Wang
- Department of Nuclear Medicine, First Medical Center, Chinese PLA General Hospital, Beijing, China
- Ruimin Wang,
| |
Collapse
|
12
|
Li Y, Liu T, Cui M. Recent development in selective Tau tracers for PET imaging in the brain. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
13
|
Rai H, Gupta S, Kumar S, Yang J, Singh SK, Ran C, Modi G. Near-Infrared Fluorescent Probes as Imaging and Theranostic Modalities for Amyloid-Beta and Tau Aggregates in Alzheimer's Disease. J Med Chem 2022; 65:8550-8595. [PMID: 35759679 DOI: 10.1021/acs.jmedchem.1c01619] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A person suspected of having Alzheimer's disease (AD) is clinically diagnosed for the presence of principal biomarkers, especially misfolded amyloid-beta (Aβ) and tau proteins in the brain regions. Existing radiotracer diagnostic tools, such as PET imaging, are expensive and have limited availability for primary patient screening and pre-clinical animal studies. To change the status quo, small-molecular near-infrared (NIR) probes have been rapidly developed, which may serve as an inexpensive, handy imaging tool to comprehend the dynamics of pathogenic progression in AD and assess therapeutic efficacy in vivo. This Perspective summarizes the biochemistry of Aβ and tau proteins and then focuses on structurally diverse NIR probes with coverages of their spectroscopic properties, binding affinity toward Aβ and tau species, and theranostic effectiveness. With the summarized information and perspective discussions, we hope that this paper may serve as a guiding tool for designing novel in vivo imaging fluoroprobes with theranostic capabilities in the future.
Collapse
Affiliation(s)
- Himanshu Rai
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Sarika Gupta
- Molecular Science Laboratory, National Institute of Immunology, New Delhi-110067, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Jian Yang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Sushil K Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| |
Collapse
|
14
|
Riley KJ, Graner BD, Veronesi MC. The tauopathies: Neuroimaging characteristics and emerging experimental therapies. J Neuroimaging 2022; 32:565-581. [PMID: 35470528 PMCID: PMC9545715 DOI: 10.1111/jon.13001] [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: 01/10/2022] [Revised: 03/10/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
The tauopathies are a heterogeneous group of neurodegenerative disorders in which the prevailing underlying disease process is intracellular deposition of abnormal misfolded tau protein. Diseases often categorized as tauopathies include progressive supranuclear palsy, chronic traumatic encephalopathy, corticobasal degeneration, and frontotemporal lobar degeneration. Tauopathies can be classified through clinical assessment, imaging findings, histologic validation, or molecular biomarkers tied to the underlying disease mechanism. Many tauopathies vary in their clinical presentation and overlap substantially in presentation, making clinical diagnosis of a specific primary tauopathy difficult. Anatomic imaging findings are also rarely specific to a single tauopathy, and when present may not manifest until well after the point at which therapy may be most impactful. Molecular biomarkers hold the most promise for patient care and form a platform upon which emerging diagnostic and therapeutic applications could be developed. One of the most exciting developments utilizing these molecular biomarkers for assessment of tau deposition within the brain is tau‐PET imaging utilizing novel ligands that specifically target tau protein. This review will discuss the background, significance, and clinical presentation of each tauopathy with additional attention to the pathologic mechanisms at the protein level. The imaging characteristics will be outlined with select examples of emerging imaging techniques. Finally, current treatment options and emerging therapies will be discussed. This is by no means a comprehensive review of the literature but is instead intended for the practicing radiologist as an overview of a rapidly evolving topic.
Collapse
Affiliation(s)
- Kalen J Riley
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brian D Graner
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael C Veronesi
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
15
|
Ni R, Nitsch RM. Recent Developments in Positron Emission Tomography Tracers for Proteinopathies Imaging in Dementia. Front Aging Neurosci 2022; 13:751897. [PMID: 35046791 PMCID: PMC8761855 DOI: 10.3389/fnagi.2021.751897] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
An early detection and intervention for dementia represent tremendous unmet clinical needs and priorities in society. A shared feature of neurodegenerative diseases causing dementia is the abnormal accumulation and spreading of pathological protein aggregates, which affect the selective vulnerable circuit in a disease-specific pattern. The advancement in positron emission tomography (PET) biomarkers has accelerated the understanding of the disease mechanism and development of therapeutics for Alzheimer's disease and Parkinson's disease. The clinical utility of amyloid-β PET and the clinical validity of tau PET as diagnostic biomarker for Alzheimer's disease continuum have been demonstrated. The inclusion of biomarkers in the diagnostic criteria has introduced a paradigm shift that facilitated the early and differential disease diagnosis and impacted on the clinical management. Application of disease-modifying therapy likely requires screening of patients with molecular evidence of pathological accumulation and monitoring of treatment effect assisted with biomarkers. There is currently still a gap in specific 4-repeat tau imaging probes for 4-repeat tauopathies and α-synuclein imaging probes for Parkinson's disease and dementia with Lewy body. In this review, we focused on recent development in molecular imaging biomarkers for assisting the early diagnosis of proteinopathies (i.e., amyloid-β, tau, and α-synuclein) in dementia and discussed future perspectives.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
| | - Roger M. Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Harada R. [Imaging of neuropathology by PET tracers]. Nihon Yakurigaku Zasshi 2022; 157:453-457. [PMID: 36328560 DOI: 10.1254/fpj.22061] [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: 06/16/2023]
Abstract
Alzheimer's disease (AD) is one of the most common causes of dementia in the world. Neurodegeneration, gliosis, and misfolded proteins such as amyloid plaques and tau tangles are neuropathological hallmarks in AD. In vivo imaging of these neuropathological lesions would be good biomarkers to understand pathophysiology as well as surrogate markers for clinical trials. We developed THK tau radiotracers including [18F]THK-5351 and tested them in humans. Validations studies identified monoamine oxidase-B (MAO-B) as the off-target binding substrate of [18F]THK-5351. Since the elevation of MAO-B, which is highly expressed in reactive astrocytes, were observed in various neurological conditions, MAO-B would be a promising target for imaging reactive astrogliosis. In fact, [18F]THK-5351 PET studies demonstrated that high tracer uptake in site susceptible regions to occur astrogliosis in various neurological disorders. However, the lack of binding selectivity affects the interpretation of PET images. Therefore, we performed lead optimization from [18F]THK-5351 generating a selective and reversible MAO-B PET tracer, [18F]SMBT-1. These translational and reverse translational studies, from the development of PET tracers to validation of PET images, led to the generation of new biomarkers. In this review, we will introduce the development of [18F]THK-5351, identification of off-target binding substrates, imaging-autopsy validations, new tracer development ([18F]SMBT-1), and finally recent clinical studies of [18F]SMBT-1.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine
| |
Collapse
|
17
|
Gao F. Integrated Positron Emission Tomography/Magnetic Resonance Imaging in clinical diagnosis of Alzheimer's disease. Eur J Radiol 2021; 145:110017. [PMID: 34826792 DOI: 10.1016/j.ejrad.2021.110017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/30/2021] [Accepted: 10/31/2021] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disease which seriously endangers the health of the aged, is the most common etiology of senile dementia. With the increasing progress of neuroimaging technology, more and more imaging methods have been applied to study Alzheimer's disease. The emergence of integrated PET/MRI (Positron Emission Tomography/Magnetic Resonance Imaging) is a major advance in multimodal molecular imaging with many advantages on the structure of resolution and contrast of image over computed tomography (CT), PET and MRI. PET/MRI is now used stepwise in neurodegenerative diseases, and also has broad prospect of application in the early diagnosis of AD. In this review, we emphatically introduce the imaging advances of AD including functional imaging and molecular imaging, the advantages of PET/MRI over other imaging methods and prospects of PET/MRI in AD clinical diagnosis, especially in early diagnosis, clinical assessment and prediction on AD.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
18
|
Fu L, Zhou Z, Liu L, Zhang J, Xie H, Zhang X, Zhu M, Wang R. Functional Abnormality Associated With Tau Deposition in Alzheimer's Disease - A Hybrid Positron Emission Tomography/MRI Study. Front Aging Neurosci 2021; 13:758053. [PMID: 34721001 PMCID: PMC8548365 DOI: 10.3389/fnagi.2021.758053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/22/2021] [Indexed: 11/21/2022] Open
Abstract
Objective: To investigate the characteristics of tau deposition and its impact on functional connectivity (FC) in Alzheimer’s disease (AD). Methods: Hybrid PET/MRI scans with [18F]-THK5317 and neuropsychological assessments were undertaken in 26 participants with AD and 19 healthy controls (HC). The standardized uptake value ratio (SUVR) of [18F]-THK5317 PET imaging was compared between the AD and HC groups. Significant clusters that revealed higher tau deposition in the AD group compared to the HC group were selected as regions of interest (ROI) for FC analysis. We evaluated the difference in the FC between the two groups for each ROI pair. The clinical and radiological characteristics were compared between the AD patients with negative FC and AD patients with positive FC for exploratory analysis. Results: The bilateral inferior lateral temporal lobe, dorsal prefrontal cortex, precuneus, posterior cingulate cortex, hippocampus, and occipital lobe showed significantly higher [18F]-THK5317 accumulation in AD patients. Decreased FC in regions with higher SUVR was observed in AD patients, and the FC strength was negatively correlated with regional SUVR. Patients with a positive FC exhibited older ages, better cognitive performances, and a lower SUVR than patients with a negative FC. Conclusions: An impact of tau deposition was observed on FC at the individual level in AD patients. Our findings suggested that the combination of tau-PET and rs-fMRI might help predict AD progression.
Collapse
Affiliation(s)
- Liping Fu
- Department of Nuclear Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Zhi Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Linwen Liu
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinming Zhang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hengge Xie
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaojun Zhang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Mingwei Zhu
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ruimin Wang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
19
|
(S)-[ 18F]THK5117 brain uptake is associated with Aβ plaques and MAO-B enzyme in a mouse model of Alzheimer's disease. Neuropharmacology 2021; 196:108676. [PMID: 34216585 DOI: 10.1016/j.neuropharm.2021.108676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022]
Abstract
The mouse model of beta-amyloid (Aβ) deposition, APP/PS1-21, exhibits high brain uptake of the tau-tracer (S)-[18F]THK5117, although no neurofibrillary tangles are present in this mouse model. For this reason we investigated (S)-[18F]THK5117 off-target binding to Aβ plaques and MAO-B enzyme in APP/PS1-21 transgenic (TG) mouse model of Aβ deposition. APP/PS1-21 TG and wild-type (WT) control mice in four different age groups (2-26 months) were imaged antemortem by positron emission tomography with (S)-[18F]THK5117, and then brain autoradiography. Additional animals were used for immunohistochemical staining and MAO-B enzyme blocking study with deprenyl pre-treatment. Regional standardized uptake value ratios for the cerebellum revealed a significant temporal increase in (S)-[18F]THK5117 uptake in aged TG, but not WT, brain. Immunohistochemical staining revealed a similar increase in Aβ plaques but not endogenous hyper-phosphorylated tau or MAO-B enzyme, and ex vivo autography showed that uptake of (S)-[18F]THK5117 co-localized with the amyloid pathology. Deprenyl hydrochloride pre-treatment reduced the binding of (S)-[18F]THK5117 in the neocortex, hippocampus, and thalamus. This study's findings suggest that increased (S)-[18F]THK5117 binding in aging APP/PS1-21 TG mice is mainly due to increasing Aβ deposition, and to a lesser extent binding to MAO-B enzyme, but not hyper-phosphorylated tau.
Collapse
|
20
|
An Update on the State of Tau Radiotracer Development: a Brief Review. Mol Imaging Biol 2021; 23:797-808. [PMID: 33987775 DOI: 10.1007/s11307-021-01612-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Evolving scientific evidence has begun to point towards hyperphosphorylated tau as a major neurotoxic component in the pathophysiological development of many major neurodegenerative conditions. In response to a need for accurate and reliable diagnosis and disease monitoring in clinical and trial settings, there has been great effort put into the development of tau radiotracers. While first-generation and second-generation radiotracers have provided a basis for assessing tau, concerns of inadequate specificity and selectivity have continued to motivate further study of these radiotracers and the development of novel radiopharmaceuticals. Given the prospective scientific and clinical value of a valid tau radiotracer, the molecular neuroimaging community must be aware of the most recent developments in the realm of tau radiotracer development. This brief review article will critically overview the most established tau radiotracers and, most importantly, concentrate on the progress of more recently developed tau radiotracers.
Collapse
|
21
|
Comprehensive review on design perspective of PET ligands based on β-amyloids, tau and neuroinflammation for diagnostic intervention of Alzheimer’s disease. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00410-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
22
|
Clinical validity of increased cortical binding of tau ligands of the THK family and PBB3 on PET as biomarkers for Alzheimer's disease in the context of a structured 5-phase development framework. Eur J Nucl Med Mol Imaging 2021; 48:2086-2096. [PMID: 33723628 PMCID: PMC8175292 DOI: 10.1007/s00259-021-05277-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/21/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE The research community has focused on defining reliable biomarkers for the early detection of the pathological hallmarks of Alzheimer's disease (AD). In 2017, the Geneva AD Biomarker Roadmap initiative adapted the framework for the systematic validation of oncological biomarkers to AD, with the aim to accelerate their development and implementation in clinical practice. The aim of this work was to assess the validation status of tau PET ligands of the THK family and PBB3 as imaging biomarkers for AD, based on the Biomarker Roadmap methodology. METHODS A panel of experts in AD biomarkers convened in November 2019 at a 2-day workshop in Geneva. The level of clinical validity of tau PET ligands of the THK family and PBB3 was assessed based on the 5-phase development framework before the meeting and discussed during the workshop. RESULTS PET radioligands of the THK family discriminate well between healthy controls and patients with AD dementia (phase 2; partly achieved) and recent evidence suggests an accurate diagnostic accuracy at the mild cognitive impairment (MCI) stage of the disease (phase 3; partly achieved). The phases 2 and 3 were considered not achieved for PBB3 since no evidence exists about the ligand's diagnostic accuracy. Preliminary evidence exists about the secondary aims of each phase for all ligands. CONCLUSION Much work remains for completing the aims of phases 2 and 3 and replicating the available evidence. However, it is unlikely that the validation process for these tracers will be completed, given the presence of off-target binding and the development of second-generation tracers with improved binding and pharmacokinetic properties.
Collapse
|
23
|
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for approximately 50% to 80% of all cases of dementia. The diagnosis of probable AD is based on clinical criteria and overlapping clinical features pose a challenge to accurate diagnosis. However, neuroimaging has been included as a biomarker in various published criteria for the diagnosis of probable AD, in the absence of a confirmatory diagnostic test during life. Advances in neuroimaging techniques and their inclusion in diagnostic and research criteria for the diagnosis of AD includes the use of positron emission tomography (PET) imaging as a biomarker in various therapeutic and prognostic studies in AD. The development and application of a range of PET tracers will allow more detailed assessment of people with AD and will improve diagnostic specificity and targeted therapy of AD. The aim of this review is to summarize current evidence on PET imaging using the non-specific tracer [18F]fluorodeoxyglucose and specific tracers that target amyloid and tau pathology in people with AD.
Collapse
Affiliation(s)
- Shailendra Mohan Tripathi
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| |
Collapse
|
24
|
Tamil Selvan S, Ravichandar R, Kanta Ghosh K, Mohan A, Mahalakshmi P, Gulyás B, Padmanabhan P. Coordination chemistry of ligands: Insights into the design of amyloid beta/tau-PET imaging probes and nanoparticles-based therapies for Alzheimer’s disease. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
van Oostveen WM, de Lange ECM. Imaging Techniques in Alzheimer's Disease: A Review of Applications in Early Diagnosis and Longitudinal Monitoring. Int J Mol Sci 2021; 22:ijms22042110. [PMID: 33672696 PMCID: PMC7924338 DOI: 10.3390/ijms22042110] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting many individuals worldwide with no effective treatment to date. AD is characterized by the formation of senile plaques and neurofibrillary tangles, followed by neurodegeneration, which leads to cognitive decline and eventually death. INTRODUCTION In AD, pathological changes occur many years before disease onset. Since disease-modifying therapies may be the most beneficial in the early stages of AD, biomarkers for the early diagnosis and longitudinal monitoring of disease progression are essential. Multiple imaging techniques with associated biomarkers are used to identify and monitor AD. AIM In this review, we discuss the contemporary early diagnosis and longitudinal monitoring of AD with imaging techniques regarding their diagnostic utility, benefits and limitations. Additionally, novel techniques, applications and biomarkers for AD research are assessed. FINDINGS Reduced hippocampal volume is a biomarker for neurodegeneration, but atrophy is not an AD-specific measure. Hypometabolism in temporoparietal regions is seen as a biomarker for AD. However, glucose uptake reflects astrocyte function rather than neuronal function. Amyloid-β (Aβ) is the earliest hallmark of AD and can be measured with positron emission tomography (PET), but Aβ accumulation stagnates as disease progresses. Therefore, Aβ may not be a suitable biomarker for monitoring disease progression. The measurement of tau accumulation with PET radiotracers exhibited promising results in both early diagnosis and longitudinal monitoring, but large-scale validation of these radiotracers is required. The implementation of new processing techniques, applications of other imaging techniques and novel biomarkers can contribute to understanding AD and finding a cure. CONCLUSIONS Several biomarkers are proposed for the early diagnosis and longitudinal monitoring of AD with imaging techniques, but all these biomarkers have their limitations regarding specificity, reliability and sensitivity. Future perspectives. Future research should focus on expanding the employment of imaging techniques and identifying novel biomarkers that reflect AD pathology in the earliest stages.
Collapse
Affiliation(s)
- Wieke M. van Oostveen
- Faculty of Science, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands;
| | - Elizabeth C. M. de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-527-6330
| |
Collapse
|
26
|
Pascoal TA, Therriault J, Benedet AL, Savard M, Lussier FZ, Chamoun M, Tissot C, Qureshi MNI, Kang MS, Mathotaarachchi S, Stevenson J, Hopewell R, Massarweh G, Soucy JP, Gauthier S, Rosa-Neto P. 18F-MK-6240 PET for early and late detection of neurofibrillary tangles. Brain 2021; 143:2818-2830. [PMID: 32671408 DOI: 10.1093/brain/awaa180] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/30/2020] [Accepted: 04/14/2020] [Indexed: 11/15/2022] Open
Abstract
Braak stages of tau neurofibrillary tangle accumulation have been incorporated in the criteria for the neuropathological diagnosis of Alzheimer's disease. It is expected that Braak staging using brain imaging can stratify living individuals according to their individual patterns of tau deposition, which may prove crucial for clinical trials and practice. However, previous studies using the first-generation tau PET agents have shown a low sensitivity to detect tau pathology in areas corresponding to early Braak histopathological stages (∼20% of cognitively unimpaired elderly with tau deposition in regions corresponding to Braak I-II), in contrast to ∼80-90% reported in post-mortem cohorts. Here, we tested whether the novel high affinity tau tangles tracer 18F-MK-6240 can better identify individuals in the early stages of tau accumulation. To this end, we studied 301 individuals (30 cognitively unimpaired young, 138 cognitively unimpaired elderly, 67 with mild cognitive impairment, 54 with Alzheimer's disease dementia, and 12 with frontotemporal dementia) with amyloid-β 18F-NAV4694, tau 18F-MK-6240, MRI, and clinical assessments. 18F-MK-6240 standardized uptake value ratio images were acquired at 90-110 min after the tracer injection. 18F-MK-6240 discriminated Alzheimer's disease dementia from mild cognitive impairment and frontotemporal dementia with high accuracy (∼85-100%). 18F-MK-6240 recapitulated topographical patterns consistent with the six hierarchical stages proposed by Braak in 98% of our population. Cognition and amyloid-β status explained most of the Braak stages variance (P < 0.0001, R2 = 0.75). No single region of interest standardized uptake value ratio accurately segregated individuals into the six topographic Braak stages. Sixty-eight per cent of the cognitively unimpaired elderly amyloid-β-positive and 37% of the cognitively unimpaired elderly amyloid-β-negative subjects displayed tau deposition, at least in the transentorhinal cortex (Braak I). Tau deposition solely in the transentorhinal cortex was associated with an elevated prevalence of amyloid-β, neurodegeneration, and cognitive impairment (P < 0.0001). 18F-MK-6240 deposition in regions corresponding to Braak IV-VI was associated with the highest prevalence of neurodegeneration, whereas in Braak V-VI regions with the highest prevalence of cognitive impairment. Our results suggest that the hierarchical six-stage Braak model using 18F-MK-6240 imaging provides an index of early and late tau accumulation as well as disease stage in preclinical and symptomatic individuals. Tau PET Braak staging using high affinity tracers has the potential to be incorporated in the diagnosis of living patients with Alzheimer's disease in the near future.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Muhammad Naveed Iqbal Qureshi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Robert Hopewell
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Gassan Massarweh
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease ResearchUnit, McGill University, Montreal, Canada.,Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Psychiatry, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Abstract
This article presents an overview of imaging agents for PET that have been applied for research and diagnostic purposes in patients affected by dementia. Classified by the target which the agents visualize, seven groups of tracers can be distinguished, namely radiopharmaceuticals for: (1) Misfolded proteins (ß-amyloid, tau, α-synuclein), (2) Neuroinflammation (overexpression of translocator protein), (3) Elements of the cholinergic system, (4) Elements of monoamine neurotransmitter systems, (5) Synaptic density, (6) Cerebral energy metabolism (glucose transport/ hexokinase), and (7) Various other proteins. This last category contains proteins involved in mechanisms underlying neuroinflammation or cognitive impairment, which may also be potential therapeutic targets. Many receptors belong to this category: AMPA, cannabinoid, colony stimulating factor 1, metabotropic glutamate receptor 1 and 5 (mGluR1, mGluR5), opioid (kappa, mu), purinergic (P2X7, P2Y12), sigma-1, sigma-2, receptor for advanced glycation endproducts, and triggering receptor expressed on myeloid cells-1, besides several enzymes: cyclooxygenase-1 and 2 (COX-1, COX-2), phosphodiesterase-5 and 10 (PDE5, PDE10), and tropomyosin receptor kinase. Significant advances in neuroimaging have been made in the last 15 years. The use of 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) for quantification of regional cerebral glucose metabolism is well-established. Three tracers for ß-amyloid plaques have been approved by the Food and Drug Administration and European Medicines Agency. Several tracers for tau neurofibrillary tangles are already applied in clinical research. Since many novel agents are in the preclinical or experimental stage of development, further advances in nuclear medicine imaging can be expected in the near future. PET studies with established tracers and tracers for novel targets may result in early diagnosis and better classification of neurodegenerative disorders and in accurate monitoring of therapy trials which involve these targets. PET data have prognostic value and may be used to assess the response of the human brain to interventions, or to select the appropriate treatment strategy for an individual patient.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sofia Marcolini
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands
| | - Peter Paul de Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands; University of Antwerp, Born-Bunge Institute, Neurochemistry and Behavior, Campus Drie Eiken, Wilrijk, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands; Ghent University, Ghent, Belgium
| |
Collapse
|
28
|
Chotipanich C, Kongthai S, Kunawudhi A, Promteangtrong C, Jantarato A. 18F-THK 5351 and 11C-PiB PET of the Thai normal brain template. ASIA OCEANIA JOURNAL OF NUCLEAR MEDICINE & BIOLOGY 2021; 9:21-30. [PMID: 33392346 DOI: 10.22038/aojnmb.2020.49623.1338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Objectives The aim of the study was to create a local normal database brain template of Thai individuals for 11C-Pittsburgh compound B (11C-PiB) and 18F-THK 5351 depositions using statistical parametric mapping (SPM) software, and to validate and optimize the established specific brain template for use in clinical practice with a highly reliability and reproducibility. Methods This prospective study was conducted in 24 healthy right-handed volunteers (13 men, 11 women; aged: 42-79 years) who underwent 18F-THK 5351 and 11C-PiB PET/CT scans. SPM was used for the 18F-THK 5351 and 11C-PiB PET/CT image analysis. All PET images were processed individually using Diffusion Tensor Image -Magnetic Resonance Imaging-weighted images (DTI-MRI images), which involved: (1) conversion of Digital Imaging and Communications in Medicine (DICOM) files into an analyzable file extension (.NIFTI) for statistical parametric mapping, (2) setting of the origin (the anterior commissure was used as the anatomical landmark), (3) re-alignment, (4) co-registration of PET with B0 (T1W) and DTI-MRI images, (5) normalization, and (6) normal verification using the Thai MRI standard. We then compared the normal PET template with the abnormal deposition area of different dementia syndromes, including Alzheimer's disease and progressive supranuclear palsy. Results This method was able to differentiate cognitively normal from Alzheimer's disease and progressive supranuclear palsy subjects. Conclusions This normal brain template was able to be integrated into clinical practice and research using PET analyses at our center.
Collapse
Affiliation(s)
- Chanisa Chotipanich
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Supaporn Kongthai
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Anchisa Kunawudhi
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | - Attapon Jantarato
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
29
|
Abstract
Pathological accumulated misfolded tau underlies various neurodegenerative diseases and associated clinical syndromes. To diagnose those diseases reliable before death or even at early stages, many different tau-specific radiotracers have been developed in the last decade to be used with positron-emission-tomography. In contrast to amyloid-β imaging, different isoforms of tau exist further complicating radiotracer development. First-generation radiotracers like [11C]PBB3, [18F]AV1451 and [18F]THK5351 have been extensively investigated in vitro and in vivo. In Alzheimer's disease (AD), high specific binding could be detected, and evidence of clinical applicability recently led to clinical approval of [18F]flortaucipir ([18F]AV1451) by the FDA. Nevertheless, absent or minor binding to non-AD tau isoforms and high off-target binding to non-tau brain structures limit the diagnostic applicability especially in non-AD tauopathies demanding further tracer development. In vitro assays and autoradiography results of next-generation radiotracers [18F]MK-6240, [18F]RO-948, [18F]PM-PBB3, [18F]GTP-1 and [18F]PI-2620 clearly indicate less off-target binding and high specific binding to tau neurofibrils. First in human studies have been conducted with promising results for all tracers in AD patients, and also some positive experience in non-AD tauopathies. Overall, larger scaled autoradiography and human studies are needed to further evaluate the most promising candidates and support future clinical approval.
Collapse
Affiliation(s)
- Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
30
|
The Imaging Features and Clinical Associations of a Novel Tau PET Tracer-18F-APN1607 in Alzheimer Disease. Clin Nucl Med 2020; 45:747-756. [PMID: 32701794 DOI: 10.1097/rlu.0000000000003164] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF THE REPORT In vivo tau PET imaging could help clarify the spatial distribution of tau deposition in Alzheimer disease (AD) and aid in the differential diagnosis of tauopathies. To date, there have been no in vivo F-APN1607 tau PET studies in patients with AD. METHODS We applied tau tracer in 12 normal controls (NCs) and 10 patients in the mild to moderate stage of probable AD. Detailed clinical information, cognitive measurements, and disease severity were documented. Regional SUV ratios (SUVRs) from F-AV-45 (florbetapir), F-APN1607 PET images, and regional gray matter (GM) atrophic ratios were calculated for further analysis. RESULTS Quantitative analyses showed significantly elevated SUVRs in the frontal, temporal, parietal, occipital lobes, anterior and posterior cingulate gyri, precuneus, and parahippocampal region (all P's < 0.01) with medium to large effect sizes (0.44-0.75). The SUVRs from F-APN1607 PET imaging showed significant correlations with the Alzheimer's Disease Assessment Scale (ADAS-cog) scores (all P's < 0.01) and strong correlation coefficients (R ranged from 0.54 to 0.68), even adjusted for age and sex effects. Finally, the SUVRs from F-APN1607 PET imaging of the parahippocampal region showed rapid saturation as the ADAS-cog scores increased, and the SUVRs of the posterior cingulate gyrus and the temporal, frontal, parietal, and occipital regions slowly increased. The combined SUVRs from amyloid, tau PET, and regional GM atrophic ratio showed regional specific patterns as the ADAS-cog scores increased. CONCLUSIONS Our findings suggest that the F-APN1607 tau tracer correlated well with cognitive changes and demonstrated the spatial pattern of amyloid, tau deposition, and GM atrophy in the progression of AD.
Collapse
|
31
|
Kelberman M, Keilholz S, Weinshenker D. What's That (Blue) Spot on my MRI? Multimodal Neuroimaging of the Locus Coeruleus in Neurodegenerative Disease. Front Neurosci 2020; 14:583421. [PMID: 33122996 PMCID: PMC7573566 DOI: 10.3389/fnins.2020.583421] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023] Open
Abstract
The locus coeruleus (LC) has long been underappreciated for its role in the pathophysiology of Alzheimer’s disease (AD), Parkinson’s disease (PD), and other neurodegenerative disorders. While AD and PD are distinct in clinical presentation, both are characterized by prodromal protein aggregation in the LC, late-stage degeneration of the LC, and comorbid conditions indicative of LC dysfunction. Many of these early studies were limited to post-mortem histological techniques due to the LC’s small size and location deep in the brainstem. Thus, there is a growing interest in utilizing in vivo imaging of the LC as a predictor of preclinical neurodegenerative processes and biomarker of disease progression. Simultaneously, neuroimaging in animal models of neurodegenerative disease holds promise for identifying early alterations to LC circuits, but has thus far been underutilized. While still in its infancy, a handful of studies have reported effects of single gene mutations and pathology on LC function in disease using various neuroimaging techniques. Furthermore, combining imaging and optogenetics or chemogenetics allows for interrogation of network connectivity in response to changes in LC activity. The purpose of this article is twofold: (1) to review what magnetic resonance imaging (MRI) and positron emission tomography (PET) have revealed about LC dysfunction in neurodegenerative disease and its potential as a biomarker in humans, and (2) to explore how animal models can be used to test hypotheses derived from clinical data and establish a mechanistic framework to inform LC-focused therapeutic interventions to alleviate symptoms and impede disease progression.
Collapse
Affiliation(s)
- Michael Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Shella Keilholz
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| |
Collapse
|
32
|
Dalton RM, Krishnan HS, Parker VS, Catanese MC, Hooker JM. Coevolution of Atomic Resolution and Whole-Brain Imaging for Tau Neurofibrillary Tangles. ACS Chem Neurosci 2020; 11:2513-2522. [PMID: 32786315 DOI: 10.1021/acschemneuro.0c00426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Neurofibrillary tangle (NFT) imaging methods at the distinct scales of atomic and whole-brain resolutions have coevolved rapidly. Linking these two areas of research provides insight into how and why certain tau radiotracers, using positron emission tomography (PET), bind selectively to certain morphological forms of the NFT fibril. In this Review, a brief history and background for each research area is presented leading to a summary of the current state of knowledge, with a synopsis of PET NFT radiotracers and an outlook for near-term research efforts. The continued integration of information provided at the level of each of these scales of resolution will catalyze the next generation of clinical imaging technique development and enhance our interpretations of them.
Collapse
Affiliation(s)
- Raeann M. Dalton
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
| | - Hema S. Krishnan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
- Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Victoria S. Parker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
- Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Mary C. Catanese
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
- Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Jacob M. Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
- Harvard Medical School, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
33
|
Kong Y, Liu C, Zhou Y, Qi J, Zhang C, Sun B, Wang J, Guan Y. Progress of RAGE Molecular Imaging in Alzheimer's Disease. Front Aging Neurosci 2020; 12:227. [PMID: 32848706 PMCID: PMC7417350 DOI: 10.3389/fnagi.2020.00227] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by senile plaques (SPs), which are caused by amyloid beta (Aβ) deposition and neurofibrillary tangles (NFTs) of abnormal hyperphosphorylated tau protein. The receptor for advanced glycation end products (RAGE) binds to advanced glycation end products deposited during vascular dysfunction. Alzheimer’s disease may occur when RAGE binds to Aβ and releases reactive oxygen species, further exacerbating Aβ deposition and eventually leading to SPs and NFTs. As it is involved in early AD, RAGE may be considered as a more potent biomarker than Aβ. Positron emission tomography provides valuable information regarding the underlying pathological processes of AD many years before the appearance of clinical symptoms. Thus, to further reveal the role of RAGE in AD pathology and for early diagnosis of AD, a tracer that targets RAGE is needed. In this review, we first describe the early diagnosis of AD and then summarize the interaction between RAGE and Aβ and Tau that is required to induce AD pathology, and finally focus on RAGE-targeting probes, highlighting the potential of RAGE to be used as an effective target. The development of RAGE probes is expected to aid in AD diagnosis and treatment.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Cuiping Liu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yinping Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jingxuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Vanhaute H, Ceccarini J, Michiels L, Koole M, Sunaert S, Lemmens R, Triau E, Emsell L, Vandenbulcke M, Van Laere K. In vivo synaptic density loss is related to tau deposition in amnestic mild cognitive impairment. Neurology 2020; 95:e545-e553. [PMID: 32493717 DOI: 10.1212/wnl.0000000000009818] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/09/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To investigate in vivo whether synaptic loss and neurofibrillary tangle load spatially overlap and correlate with clinical symptoms in patients with amnestic mild cognitive impairment (aMCI). METHODS In this cross-sectional study, 10 patients with aMCI and 10 healthy controls underwent triple PET-MRI with 11C-UCB-J (synaptic vesicle protein 2A), 18F-MK-6240 (tau deposition), and 11C-Pittsburgh compound B (β-amyloid) and neuropsychological assessment. Gray matter atrophy was assessed by voxel-based morphometry with T1-weighted MRIs. Voxel-wise and volume-of-interest analyses were conducted on PET data. The interrelationship of synaptic density and tau deposition was investigated. We also investigated correlations of 18F-MK-6240 and 11C-UCB-J binding with cognitive performance. RESULTS Compared to controls, patients with aMCI showed a decreased 11C-UCB-J binding mainly in substructures of the medial temporal lobe (MTL; 48%-51%, p cluster = 0.02). Increased 18F-MK6240 binding in the same region was observed (42%-44%, p cluster = 0.0003), spreading to association cortices. In the MTL, higher 18F-MK-6240 binding inversely related to lower 11C-UCB-J binding (p = 0.02, r = -0.76). Decreased performance on cognitive tests was associated with both increased 18F-MK-6240 and decreased 11C-UCB-J binding in the hippocampus (p < 0.01, r > 0.7), although in a multivariate analysis only 18F-MK-6240 binding was significantly related to cognitive performance. CONCLUSIONS Patients with aMCI have high tau deposition and synaptic density loss mainly in key regions known to be involved in early cognitive impairment, indicating that these are interrelated in the MTL, while tau binding had already spread toward association cortices. Longitudinal data are needed to provide further insight into the temporal aspects of this relationship.
Collapse
Affiliation(s)
- Heleen Vanhaute
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium.
| | - Jenny Ceccarini
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Laura Michiels
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Michel Koole
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Stefan Sunaert
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Robin Lemmens
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Eric Triau
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Louise Emsell
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Mathieu Vandenbulcke
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| | - Koen Van Laere
- From the Division of Nuclear Medicine (H.V., J.C., M.K., K.V.L.) and Department of Neurology (L.M., R.L.), University Hospitals Leuven; Nuclear Medicine and Molecular Imaging (H.V., J.C., M.K., S.S., L.E., K.V.L.) and Translational MRI (S.S., L.E.), Department of Imaging and Pathology, and Department of Geriatric Psychiatry (H.V., L.E., M.V.), University Psychiatric Centre, Laboratory for Neurobiology (L.M., R.L.), KU Leuven; and Center for Brain and Disease Research (L.M., R.L.), VIB-KU Leuven, Belgium. Dr. Triau is in private practice in Leuven, Belgium
| |
Collapse
|
35
|
Gyasi YI, Pang YP, Li XR, Gu JX, Cheng XJ, Liu J, Xu T, Liu Y. Biological applications of near infrared fluorescence dye probes in monitoring Alzheimer’s disease. Eur J Med Chem 2020; 187:111982. [DOI: 10.1016/j.ejmech.2019.111982] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/03/2019] [Accepted: 12/16/2019] [Indexed: 01/10/2023]
|
36
|
Beaurain M, Salabert AS, Ribeiro MJ, Arlicot N, Damier P, Le Jeune F, Demonet JF, Payoux P. Innovative Molecular Imaging for Clinical Research, Therapeutic Stratification, and Nosography in Neuroscience. Front Med (Lausanne) 2019; 6:268. [PMID: 31828073 PMCID: PMC6890558 DOI: 10.3389/fmed.2019.00268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/01/2019] [Indexed: 01/06/2023] Open
Abstract
Over the past few decades, several radiotracers have been developed for neuroimaging applications, especially in PET. Because of their low steric hindrance, PET radionuclides can be used to label molecules that are small enough to cross the blood brain barrier, without modifying their biological properties. As the use of 11C is limited by its short physical half-life (20 min), there has been an increasing focus on developing tracers labeled with 18F for clinical use. The first such tracers allowed cerebral blood flow and glucose metabolism to be measured, and the development of molecular imaging has since enabled to focus more closely on specific targets such as receptors, neurotransmitter transporters, and other proteins. Hence, PET and SPECT biomarkers have become indispensable for innovative clinical research. Currently, the treatment options for a number of pathologies, notably neurodegenerative diseases, remain only supportive and symptomatic. Treatments that slow down or reverse disease progression are therefore the subject of numerous studies, in which molecular imaging is proving to be a powerful tool. PET and SPECT biomarkers already make it possible to diagnose several neurological diseases in vivo and at preclinical stages, yielding topographic, and quantitative data about the target. As a result, they can be used for assessing patients' eligibility for new treatments, or for treatment follow-up. The aim of the present review was to map major innovative radiotracers used in neuroscience, and explain their contribution to clinical research. We categorized them according to their target: dopaminergic, cholinergic or serotoninergic systems, β-amyloid plaques, tau protein, neuroinflammation, glutamate or GABA receptors, or α-synuclein. Most neurological disorders, and indeed mental disorders, involve the dysfunction of one or more of these targets. Combinations of molecular imaging biomarkers can afford us a better understanding of the mechanisms underlying disease development over time, and contribute to early detection/screening, diagnosis, therapy delivery/monitoring, and treatment follow-up in both research and clinical settings.
Collapse
Affiliation(s)
- Marie Beaurain
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Anne-Sophie Salabert
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Maria Joao Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Philippe Damier
- Inserm U913, Neurology Department, University Hospital, Nantes, France
| | | | - Jean-François Demonet
- Leenards Memory Centre, Department of Clinical Neuroscience, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pierre Payoux
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| |
Collapse
|
37
|
AD molecular: Imaging tau aggregates with positron emissions tomography. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 165:107-138. [PMID: 31481160 DOI: 10.1016/bs.pmbts.2019.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pathologic aggregates of tau protein are observed in several neurodegenerative diseases and are used to diagnose and stage disease postmortem. Recent advances in positron emission tomography radioligands allow for the detection of aggregated tau proteins in living persons. This chapter describes the development and characterization of several positron emission tomography radioligands used to detect tau pathophysiology in vivo, and how these ligands are being used in clinical aging and neurodegenerative disease research with a focus on imaging tau aggregates in Alzheimer's disease.
Collapse
|
38
|
Jeon S, Kang JM, Seo S, Jeong HJ, Funck T, Lee SY, Park KH, Lee YB, Yeon BK, Ido T, Okamura N, Evans AC, Na DL, Noh Y. Topographical Heterogeneity of Alzheimer's Disease Based on MR Imaging, Tau PET, and Amyloid PET. Front Aging Neurosci 2019; 11:211. [PMID: 31481888 PMCID: PMC6710378 DOI: 10.3389/fnagi.2019.00211] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/26/2019] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) patients are known to have heterogeneous clinical presentation and pathologic patterns. We hypothesize that AD dementia can be categorized into subtypes based on multimodal imaging biomarkers such as magnetic resonance imaging (MRI), tau positron emission tomography (PET), and amyloid PET. We collected 3T MRI, 18F-THK5351 PET, and 18F-flutemetamol (FLUTE) PET data from 83 patients with AD dementia [Clinical Dementia Rating (CDR) ≤1] and 60 normal controls (NC), and applied surface-based analyses to measure cortical thickness, THK5351 standardized uptake value ratio (SUVR) and FLUTE SUVR for each participant. For the patient group, we performed an agglomerative hierarchical clustering analysis using the three multimodal imaging features on the vertices (n = 3 × 79,950). The identified AD subtypes were compared to NC using general linear models adjusting for age, sex, and years of education. We mapped the effect size within significant cortical regions reaching a corrected p-vertex <0.05 (random field theory). Our surface-based multimodal framework has revealed three distinct subtypes among AD patients: medial temporal-dominant subtype (MT, n = 44), parietal-dominant subtype (P, n = 19), and diffuse atrophy subtype (D, n = 20). The topography of cortical atrophy and THK5351 retention differentiates between the three subtypes. In the case of FLUTE, three subtypes did not show distinct topographical differences, although cortical composite retention was significantly higher in the P type than in the MT type. These three subtypes also differed in demographic and clinical features. In conclusion, AD patients may be clustered into three subtypes with distinct topographical features of cortical atrophy and tau deposition, although amyloid deposition may not differ across the subtypes in terms of topography.
Collapse
Affiliation(s)
- Seun Jeon
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jae Myeong Kang
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Seongho Seo
- Department of Neuroscience, Gachon University College of Medicine, Incheon, South Korea
| | - Hye Jin Jeong
- Neuroscience Research Institute, Gachon University, Incheon, South Korea
| | - Thomas Funck
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Sang-Yoon Lee
- Department of Neuroscience, Gachon University College of Medicine, Incheon, South Korea
| | - Kee Hyung Park
- Department of Neurology, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Yeong-Bae Lee
- Department of Neurology, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Byeong Kil Yeon
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Tatsuo Ido
- Neuroscience Research Institute, Gachon University, Incheon, South Korea
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Alan C Evans
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Young Noh
- Department of Neurology, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea.,Department of Health Science and Technology, GAIHST, Gachon University, Incheon, South Korea
| |
Collapse
|
39
|
Leinonen V, Rauramaa T, Johansson J, Bottelbergs A, Tesseur I, van der Ark P, Pemberton D, Koivisto AM, Jääskeläinen JE, Hiltunen M, Herukka SK, Blennow K, Zetterberg H, Jokinen P, Rokka J, Helin S, Haaparanta-Solin M, Solin O, Okamura N, Kolb HC, Rinne JO. S-[18F]THK-5117-PET and [11C]PIB-PET Imaging in Idiopathic Normal Pressure Hydrocephalus in Relation to Confirmed Amyloid-β Plaques and Tau in Brain Biopsies. J Alzheimers Dis 2019; 64:171-179. [PMID: 29865068 DOI: 10.3233/jad-180071] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Detection of pathological tau aggregates could facilitate clinical diagnosis of Alzheimer's disease (AD) and monitor drug effects in clinical trials. S-[18F]THK-5117 could be a potential tracer to detect pathological tau deposits in brain. However, no previous study have correlated S-[18F]THK-5117 uptake in PET with brain biopsy verified tau pathology in vivo. OBJECTIVE Here we aim to evaluate the association between cerebrospinal fluid (CSF) AD biomarkers, S-[18F]THK-5117, and [11C]PIB PET against tau and amyloid lesions in brain biopsy. METHODS Fourteen patients with idiopathic normal pressure hydrocephalus (iNPH) with previous shunt surgery including right frontal cortical brain biopsy and CSF Aβ1 - 42, total tau, and P-tau181 measures, underwent brain MRI, [11C]PIB PET, and S-[18F]THK-5117 PET imaging. RESULTS Seven patients had amyloid-β (Aβ, 4G8) plaques, two both Aβ and phosphorylated tau (Pτ, AT8) and one only Pτ in biopsy. As expected, increased brain biopsy Aβ was well associated with higher [11C]PIB uptake in PET. However, S-[18F]THK-5117 uptake did not show any statistically significant correlation with either brain biopsy Pτ or CSF P-tau181 or total tau. CONCLUSIONS S-[18F]THK-5117 lacked clear association with neuropathologically verified tau pathology in brain biopsy probably, at least partially, due to off-target binding. Further studies with larger samples of patients with different tau tracers are urgently needed. The detection of simultaneous Aβ and tau pathology in iNPH is important since that may indicate poorer and especially shorter response for CSF shunt surgery compared with no pathology.
Collapse
Affiliation(s)
- Ville Leinonen
- Institute of Clinical Medicine - Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland.,Unit of ClinicalNeuroscience, Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Tuomas Rauramaa
- Institute of Clinical Medicine - Pathology, University of Eastern Finland andDepartment of Pathology, Kuopio University Hospital, Kuopio, Finland
| | | | - Astrid Bottelbergs
- Janssen Research and Development, A Division of Janssen Pharmaceutica, Beerse, Belgium
| | - Ina Tesseur
- Janssen Research and Development, A Division of Janssen Pharmaceutica, Beerse, Belgium
| | - Peter van der Ark
- Janssen Research and Development, A Division of Janssen Pharmaceutica, Beerse, Belgium
| | - Darrel Pemberton
- Janssen Research and Development, A Division of Janssen Pharmaceutica, Beerse, Belgium
| | - Anne M Koivisto
- Institute of Clinical Medicine - Neurology, Universityof Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Juha E Jääskeläinen
- Institute of Clinical Medicine - Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Clinical Medicine - Neurology, Universityof Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland.,Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine - Neurology, Universityof Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska Academy Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska Academy Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Department of MolecularNeuroscience, Institute of Neurology, University College London, Queen, Square, UK.,UK DementiaResearch Institute, London, UK
| | - Pekka Jokinen
- Turku PET Centre, University of Turku, Turku, Finland.,Department of Neurosurgery, Turku University Hospital, Turku, Finland
| | - Johanna Rokka
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland.,Athinoula A. Martinos Center, Department of Radiology, Massachusetts GeneralHospital, Harvard Medical School, Charlestown, MA, USA
| | - Semi Helin
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Merja Haaparanta-Solin
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Olof Solin
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland.,Department of Chemistry, University of Turku, Turku, Finland.,Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | | | | | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku, Finland.,Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| |
Collapse
|
40
|
Abstract
Purpose of Review Abnormal accumulation of tau protein is the main hallmark of tauopathies and is closely associated with neurodegeneration and cognitive impairment, whereas the advance in PET imaging provides a non-invasive detection of tau inclusions in the brain. In this review, we discuss the potential of PET imaging as a biomarker in tauopathies, the latest development of novel tau tracers with new clinical information that has been disclosed, and the opportunities for improving diagnosis and designing clinical trials in the future. Recent Findings In recent years, several first-generation tau PET tracers including [11C]PBB3, [18F]THK-5117, [18F]THK-5351 and [18F]AV-1451 have been developed and succeeded in imaging neurofibrillary pathology in vivo. Due to the common off-target binding and subcortical white matter uptake seen in the first-generation tracers, several research institutes and pharmaceutical companies have been working on developing second-generation tau PET tracers which exhibit higher binding affinity and selectivity. Summary Tau PET imaging is promising to serve as a biomarker to support differential diagnosis and monitor disease progression in many neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi Ting Wang
- Neurology Imaging Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Paul Edison
- Neurology Imaging Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK. .,Cardiff University, Cardiff, CF10 3AT, UK.
| |
Collapse
|
41
|
Wilson H, Pagano G, Politis M. Dementia spectrum disorders: lessons learnt from decades with PET research. J Neural Transm (Vienna) 2019; 126:233-251. [PMID: 30762136 PMCID: PMC6449308 DOI: 10.1007/s00702-019-01975-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
The dementia spectrum encompasses a range of disorders with complex diagnosis, pathophysiology and limited treatment options. Positron emission tomography (PET) imaging provides insights into specific neurodegenerative processes underlying dementia disorders in vivo. Here we focus on some of the most common dementias: Alzheimer's disease, Parkinsonism dementias including Parkinson's disease with dementia, dementia with Lewy bodies, progressive supranuclear palsy and corticobasal syndrome, and frontotemporal lobe degeneration. PET tracers have been developed to target specific proteinopathies (amyloid, tau and α-synuclein), glucose metabolism, cholinergic system and neuroinflammation. Studies have shown distinct imaging abnormalities can be detected early, in some cases prior to symptom onset, allowing disease progression to be monitored and providing the potential to predict symptom onset. Furthermore, advances in PET imaging have identified potential therapeutic targets and novel methods to accurately discriminate between different types of dementias in vivo. There are promising imaging markers with a clinical application on the horizon, however, further studies are required before they can be implantation into clinical practice.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK
| | - Gennaro Pagano
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK.
| |
Collapse
|
42
|
Evaluation of the Feasibility of Screening Tau Radiotracers Using an Amyloid Biomathematical Screening Methodology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2019; 2018:6287913. [PMID: 30662517 PMCID: PMC6314003 DOI: 10.1155/2018/6287913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/06/2018] [Accepted: 11/21/2018] [Indexed: 11/17/2022]
Abstract
The purpose of this study is to evaluate the feasibility of extending a previously developed amyloid biomathematical screening methodology to support the screening of tau radiotracers during compound development. 22 tau-related PET radiotracers were investigated. For each radiotracer, in silico MLogP, V x, and in vitro K D were input into the model to predict the in vivo K 1, k 2, and BPND under healthy control (HC), mild cognitive impaired (MCI), and Alzheimer's disease (AD) conditions. These kinetic parameters were used to simulate the time activity curves (TACs) in the target regions of HC, MCI, and AD and a reference region. Standardized uptake value ratios (SUVR) were determined from the integrated area under the TACs of the target region over the reference region within a default time window of 90-110 min. The predicted K 1, k 2, and BPND values were compared with the clinically observed values. The TACs and SUVR distributions were also simulated with population variations and noise. Finally, the clinical usefulness index (CUI) ranking was compared with clinical comparison results. The TACs and SUVR distributions differed for tau radiotracers with lower tau selectivity. The CUI values ranged from 0.0 to 16.2, with 6 out of 9 clinically applied tau radiotracers having CUI values higher than the recommend CUI value of 3.0. The differences between the clinically observed TACs and SUVR results showed that the evaluation of the clinical usefulness of tau radiotracer based on single target binding could not fully reflect in vivo tau binding. The screening methodology requires further study to improve the accuracy of screening tau radiotracers. However, the higher CUI rankings of clinically applied tau radiotracers with higher signal-to-noise ratio supported the use of the screening methodology in radiotracer development by allowing comparison of candidate radiotracers with clinically applied radiotracers based on SUVR, with respect to binding to a single target.
Collapse
|
43
|
Longitudinal tau and metabolic PET imaging in relation to novel CSF tau measures in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2019; 46:1152-1163. [PMID: 30610252 PMCID: PMC6451715 DOI: 10.1007/s00259-018-4242-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/11/2018] [Indexed: 12/02/2022]
Abstract
Purpose Studies comparing CSF and PET tau biomarkers have included only commercial CSF assays examining specific phosphorylation sites (e.g. threonine 181, P-tau181p) and mid-domain tau (i.e. total tau, T-tau). Moreover, these studies did not examine CSF tau levels in relation to cerebral glucose metabolism. We thus aimed to examine CSF tau measures, using both commercial and novel assays, in relation to [18F]THK5317 (tau) and [18F]FDG PET (glucose metabolism). Methods Fourteen Alzheimer’s disease (AD) patients (seven prodromal, seven dementia) underwent [18F]THK5317 and [18F]FDG PET studies, with follow-up performed in ten subjects (six prodromal, four dementia) after 17 months. In addition to commercial assays, novel measures capturing N-terminus+mid-domain (tau N-Mid) and C-terminally truncated (tau-368) fragments were included. Results While the levels of all forms of CSF tau were found to be inversely associated with baseline [18F]FDG uptake, associations with baseline [18F]THK5317 uptake varied in relation to the degree of isocortical hypometabolism ([18F]FDG SUVR). Changes in the levels of the novel CSF markers tracked longitudinal changes in tracer uptake better than changes in P-tau181p and T-tau levels, and improved concordance with dichotomized regional [18F]THK5317 measures. Conclusion Our findings suggest that neurodegeneration may modulate the relationship between CSF and PET tau biomarkers, and that, by comparison to P-tau181p and T-tau, tau-368 and tau N-Mid may better capture tau pathology and synaptic impairment. Electronic supplementary material The online version of this article (10.1007/s00259-018-4242-6) contains supplementary material, which is available to authorized users.
Collapse
|
44
|
Leuzy A, Chiotis K, Lemoine L, Gillberg PG, Almkvist O, Rodriguez-Vieitez E, Nordberg A. Tau PET imaging in neurodegenerative tauopathies-still a challenge. Mol Psychiatry 2019; 24:1112-1134. [PMID: 30635637 PMCID: PMC6756230 DOI: 10.1038/s41380-018-0342-8] [Citation(s) in RCA: 390] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/19/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
The accumulation of pathological misfolded tau is a feature common to a collective of neurodegenerative disorders known as tauopathies, of which Alzheimer's disease (AD) is the most common. Related tauopathies include progressive supranuclear palsy (PSP), corticobasal syndrome (CBS), Down's syndrome (DS), Parkinson's disease (PD), and dementia with Lewy bodies (DLB). Investigation of the role of tau pathology in the onset and progression of these disorders is now possible due the recent advent of tau-specific ligands for use with positron emission tomography (PET), including first- (e.g., [18F]THK5317, [18F]THK5351, [18F]AV1451, and [11C]PBB3) and second-generation compounds [namely [18F]MK-6240, [18F]RO-948 (previously referred to as [18F]RO69558948), [18F]PI-2620, [18F]GTP1, [18F]PM-PBB3, and [18F]JNJ64349311 ([18F]JNJ311) and its derivative [18F]JNJ-067)]. In this review we describe and discuss findings from in vitro and in vivo studies using both initial and new tau ligands, including their relation to biomarkers for amyloid-β and neurodegeneration, and cognitive findings. Lastly, methodological considerations for the quantification of in vivo ligand binding are addressed, along with potential future applications of tau PET, including therapeutic trials.
Collapse
Affiliation(s)
- Antoine Leuzy
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden ,0000 0000 9241 5705grid.24381.3cTheme Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Laetitia Lemoine
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Per-Göran Gillberg
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ove Almkvist
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden ,0000 0004 1936 9377grid.10548.38Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Elena Rodriguez-Vieitez
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden. .,Theme Aging, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
45
|
Hopewell R, Ross K, Kostikov A, Pascoal TA, Alberti T, Lacatus-Samoila M, Soucy JP, Bennacef I, Kobayashi E, Kang MS, Rosa-Neto P, Massarweh G. A simplified radiosynthesis of [18
F]MK-6240 for tau PET imaging. J Labelled Comp Radiopharm 2018; 62:109-114. [DOI: 10.1002/jlcr.3695] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Robert Hopewell
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Karen Ross
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Alexey Kostikov
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Tharick A. Pascoal
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | - Thais Alberti
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | | | - Jean-Paul Soucy
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Idriss Bennacef
- Translational Biomarkers; Merck & Co, Inc; West Point Pennsylvania USA
| | - Eliane Kobayashi
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Min Su Kang
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | - Pedro Rosa-Neto
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | - Gassan Massarweh
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| |
Collapse
|
46
|
Abstract
The past decade has seen tremendous efforts in biomarker discovery and validation for neurodegenerative diseases. The source and type of biomarkers has continued to grow for central nervous system diseases, from biofluid-based biomarkers (blood or cerebrospinal fluid (CSF)), to nucleic acids, tissue, and imaging. While DNA remains a predominant biomarker used to identify familial forms of neurodegenerative diseases, various types of RNA have more recently been linked to familial and sporadic forms of neurodegenerative diseases during the past few years. Imaging approaches continue to evolve and are making major contributions to target engagement and early diagnostic biomarkers. Incorporation of biomarkers into drug development and clinical trials for neurodegenerative diseases promises to aid in the development and demonstration of target engagement and drug efficacy for neurologic disorders. This review will focus on recent advancements in developing biomarkers for clinical utility in Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
| | - Robert Bowser
- Iron Horse Diagnostics, Inc., Scottsdale, AZ, 85255, USA.
- Divisions of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ, 85013, USA.
| |
Collapse
|
47
|
Harada R, Okamura N, Furumoto S, Yanai K. Imaging Protein Misfolding in the Brain Using β-Sheet Ligands. Front Neurosci 2018; 12:585. [PMID: 30186106 PMCID: PMC6110819 DOI: 10.3389/fnins.2018.00585] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases characterized by pathological protein accumulation in cells are termed “proteinopathies.” Although various protein aggregates share cross-β-sheet structures, actual conformations vary among each type of protein deposit. Recent progress in the development of radiotracers for positron emission tomography (PET) has enabled the visualization of protein aggregates in living brains. Amyloid PET tracers have been developed, and are widely used for the diagnosis of Alzheimer’s disease and non-invasive assessment of amyloid burden in clinical trials of anti-dementia drugs. Furthermore, several tau PET tracers have been successfully developed and used in the clinical studies. However, recent studies have identified the presence of off-target binding of radiotracers in areas of tau deposition, suggesting that concomitant neuroinflammatory changes might affect tracer binding. In contrast to amyloid and tau PET, there are no established tracers for imaging Lewy bodies in the human brain. In this review, we describe lessons learned from the development of PET tracers and discuss the future direction of tracer development for protein misfolding diseases.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| |
Collapse
|
48
|
Valotassiou V, Malamitsi J, Papatriantafyllou J, Dardiotis E, Tsougos I, Psimadas D, Alexiou S, Hadjigeorgiou G, Georgoulias P. SPECT and PET imaging in Alzheimer’s disease. Ann Nucl Med 2018; 32:583-593. [PMID: 30128693 DOI: 10.1007/s12149-018-1292-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Varvara Valotassiou
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece.
| | - Julia Malamitsi
- Medical Physics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Ioannis Tsougos
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - Dimitrios Psimadas
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - Sotiria Alexiou
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - George Hadjigeorgiou
- Neurology Department, University Hospital of Larissa, Thessaly, Greece
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Greece
| | - Panagiotis Georgoulias
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| |
Collapse
|
49
|
Li C, Takahashi T, Shrestha T, Kinoshita E, Matsubara T, Matsumoto M, Maruyama H. 4',6-Diamidino-2-Phenylindole Distinctly Labels Tau Deposits. J Histochem Cytochem 2018; 66:737-751. [PMID: 30106598 DOI: 10.1369/0022155418793600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tau deposits have distinct biochemical characteristics and vary morphologically based on identification with tau antibodies and several chemical dyes. Here, we report 4',6-diamidino-2-phenylindole (DAPI)-positivity of tau deposits. Furthermore, we investigated the cause for this positivity. DAPI was positive in 3R/4R (3-repeat/4-repeat) tau deposits in Alzheimer's disease, myotonic dystrophy, and neurodegeneration with brain iron accumulation, and in 4R tau deposits in corticobasal degeneration, but negative in 4R tau deposits in frontotemporal dementia with parkinsonism-17 and progressive supranuclear palsy. The peak emission wavelength of DAPI after binding to a tau deposit was similar to that after binding to a nucleus. This DAPI-positivity was conspicuous at the optimum concentration of 2 μg/ml. DAPI-positivity was diminished after formic acid treatment, but preserved after nucleic acid elimination and phosphate moiety blocking. Our results suggest that staining with 2 μg/ml DAPI is a common but useful tool to differentially detect tau deposits in various tauopathies.
Collapse
Affiliation(s)
- Chengyu Li
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tetsuya Takahashi
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tejashwi Shrestha
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Eiji Kinoshita
- Department of Functional Molecular Science, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyasu Matsubara
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Masayasu Matsumoto
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan.,Sakai City Medical Center, Sakai, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
50
|
|