1
|
Wongso H, Harada R, Furumoto S. Current Progress and Future Directions in Non-Alzheimer's Disease Tau PET Tracers. ACS Chem Neurosci 2025; 16:111-127. [PMID: 39762194 DOI: 10.1021/acschemneuro.4c00319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) and non-AD tauopathies are dominant public health issues driven by several factors, especially in the aging population. The discovery of first-generation radiotracers, including [18F]FDDNP, [11C]PBB3, [18F]flortaucipir, and the [18F]THK series, for the in vivo detection of tauopathies has marked a significant breakthrough in the fields of neuroscience and radiopharmaceuticals, creating a robust new category of labeled compounds: tau positron emission tomography (PET) tracers. Subsequently, other tau PET tracers with improved binding properties have been developed using various chemical scaffolds to target the three-repeat/four-repeat (3R/4R) tau folds in AD. In 2020, [18F]flortaucipir was approved by the U.S. Food and Drug Administration for PET imaging of tau pathology in adult patients with cognitive deficits undergoing evaluation for AD. Despite remarkable progress in the development of AD tau PET tracers, imaging agents for rare non-AD tauopathies (4R tauopathies [predominantly expressing a 4R tau isoform], involved in progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, and globular glial tauopathy, and 3R tauopathies [predominantly expressing a 3R tau isoform], such as Pick's disease) remain substantially underdeveloped. In this review, we discuss recent progress in tau PET tracer development, with particular emphasis on clinically validated tracers for AD and their potential use for non-AD tauopathies. Additionally, we highlight the critical need for further development of tau PET tracers specifically designed for non-AD tauopathies, an area that remains significantly underexplored despite its importance in advancing the understanding and diagnosis of these disorders.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Ryuichi Harada
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Shozo Furumoto
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
2
|
Kumar R, Waisberg E, Ong J, Paladugu P, Amiri D, Saintyl J, Yelamanchi J, Nahouraii R, Jagadeesan R, Tavakkoli A. Artificial Intelligence-Based Methodologies for Early Diagnostic Precision and Personalized Therapeutic Strategies in Neuro-Ophthalmic and Neurodegenerative Pathologies. Brain Sci 2024; 14:1266. [PMID: 39766465 PMCID: PMC11674895 DOI: 10.3390/brainsci14121266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Advancements in neuroimaging, particularly diffusion magnetic resonance imaging (MRI) techniques and molecular imaging with positron emission tomography (PET), have significantly enhanced the early detection of biomarkers in neurodegenerative and neuro-ophthalmic disorders. These include Alzheimer's disease, Parkinson's disease, multiple sclerosis, neuromyelitis optica, and myelin oligodendrocyte glycoprotein antibody disease. This review highlights the transformative role of advanced diffusion MRI techniques-Neurite Orientation Dispersion and Density Imaging and Diffusion Kurtosis Imaging-in identifying subtle microstructural changes in the brain and visual pathways that precede clinical symptoms. When integrated with artificial intelligence (AI) algorithms, these techniques achieve unprecedented diagnostic precision, facilitating early detection of neurodegeneration and inflammation. Additionally, next-generation PET tracers targeting misfolded proteins, such as tau and alpha-synuclein, along with inflammatory markers, enhance the visualization and quantification of pathological processes in vivo. Deep learning models, including convolutional neural networks and multimodal transformers, further improve diagnostic accuracy by integrating multimodal imaging data and predicting disease progression. Despite challenges such as technical variability, data privacy concerns, and regulatory barriers, the potential of AI-enhanced neuroimaging to revolutionize early diagnosis and personalized treatment in neurodegenerative and neuro-ophthalmic disorders is immense. This review underscores the importance of ongoing efforts to validate, standardize, and implement these technologies to maximize their clinical impact.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA; (R.K.); (J.S.)
| | - Ethan Waisberg
- Department of Clinical Neurosciences, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK;
| | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St, Ann Arbor, MI 48105, USA
| | - Phani Paladugu
- Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut St, Philadelphia, PA 19107, USA;
- Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Dylan Amiri
- Department of Biology, University of Miami, 1301 Memorial Dr, Coral Gables, FL 33146, USA;
- Mecklenburg Neurology Group, 3541 Randolph Rd #301, Charlotte, NC 28211, USA;
| | - Jeremy Saintyl
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA; (R.K.); (J.S.)
| | - Jahnavi Yelamanchi
- Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY 11201, USA;
| | - Robert Nahouraii
- Mecklenburg Neurology Group, 3541 Randolph Rd #301, Charlotte, NC 28211, USA;
| | - Ram Jagadeesan
- Whiting School of Engineering, Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA;
| | - Alireza Tavakkoli
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, 1664 N Virginia St, Reno, NV 89557, USA;
| |
Collapse
|
3
|
Strobel J, Müller HP, Ludolph AC, Beer AJ, Sollmann N, Kassubek J. New Perspectives in Radiological and Radiopharmaceutical Hybrid Imaging in Progressive Supranuclear Palsy: A Systematic Review. Cells 2023; 12:2776. [PMID: 38132096 PMCID: PMC10742083 DOI: 10.3390/cells12242776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disease characterized by four-repeat tau deposition in various cell types and anatomical regions, and can manifest as several clinical phenotypes, including the most common phenotype, Richardson's syndrome. The limited availability of biomarkers for PSP relates to the overlap of clinical features with other neurodegenerative disorders, but identification of a growing number of biomarkers from imaging is underway. One way to increase the reliability of imaging biomarkers is to combine different modalities for multimodal imaging. This review aimed to provide an overview of the current state of PSP hybrid imaging by combinations of positron emission tomography (PET) and magnetic resonance imaging (MRI). Specifically, combined PET and MRI studies in PSP highlight the potential of [18F]AV-1451 to detect tau, but also the challenge in differentiating PSP from other neurodegenerative diseases. Studies over the last years showed a reduced synaptic density in [11C]UCB-J PET, linked [11C]PK11195 and [18F]AV-1451 markers to disease progression, and suggested the potential role of [18F]RO948 PET for identifying tau pathology in subcortical regions. The integration of quantitative global and regional gray matter analysis by MRI may further guide the assessment of reduced cortical thickness or volume alterations, and diffusion MRI could provide insight into microstructural changes and structural connectivity in PSP. Challenges in radiopharmaceutical biomarkers and hybrid imaging require further research targeting markers for comprehensive PSP diagnosis.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, University Hospital Ulm, 89081 Ulm, Germany;
| | - Hans-Peter Müller
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
| | - Albert C. Ludolph
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
- German Center for Neurodegenerative Diseases (DZNE), Ulm University, 89081 Ulm, Germany
| | - Ambros J. Beer
- Department of Nuclear Medicine, University Hospital Ulm, 89081 Ulm, Germany;
| | - Nico Sollmann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, 89081 Ulm, Germany;
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Jan Kassubek
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
- German Center for Neurodegenerative Diseases (DZNE), Ulm University, 89081 Ulm, Germany
| |
Collapse
|
4
|
Aramadaka S, Mannam R, Sankara Narayanan R, Bansal A, Yanamaladoddi VR, Sarvepalli SS, Vemula SL. Neuroimaging in Alzheimer's Disease for Early Diagnosis: A Comprehensive Review. Cureus 2023; 15:e38544. [PMID: 37273363 PMCID: PMC10239271 DOI: 10.7759/cureus.38544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, affecting roughly half of those over the age of 85. We briefly discussed the risk factors, epidemiology, and treatment options for AD. The development of therapeutic therapies operating very early in the disease cascade has been spurred by the realization that the disease process begins at least a decade or more before the manifestation of symptoms. Thus, the clinical significance of early diagnosis was emphasized. Using various keywords, a literature search was carried out using PubMed and other databases. For inclusion, pertinent articles were chosen and reviewed. This article has reviewed different neuroimaging techniques that are considered advanced tools to aid in establishing a diagnosis and highlighted the advantages as well as disadvantages of those techniques. Besides, the prevalence of several in vivo biomarkers aided in discriminating affected individuals from healthy controls in the early stages of the disease. Each imaging method has its advantages and disadvantages, hence no single imaging approach can be the optimum modality for diagnosis. This article also commented on a better approach to using these techniques to increase the likelihood of an early diagnosis.
Collapse
Affiliation(s)
| | - Raam Mannam
- Research, Narayana Medical College, Nellore, IND
| | | | - Arpit Bansal
- Research, Narayana Medical College, Nellore, IND
| | | | | | | |
Collapse
|
5
|
Gholampour M, Seradj H, Sakhteman A. Structure-Selectivity Relationship Prediction of Tau Imaging Tracers Using Machine Learning-Assisted QSAR Models and Interaction Fingerprint Map. ACS Chem Neurosci 2023. [PMID: 37037183 DOI: 10.1021/acschemneuro.3c00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023] Open
Abstract
Protein aggregates composed of tau fibrils are major pathologic findings in different tauopathies. An ideal agent for imaging tau fibrils must be highly selective. The molecular basis for the binding of current available compounds to tau aggregates is not well understood. Herein, we provide insights into previously studied positron emission tomography tracers using various computational methods, including machine learning-based quantitative structure-activity relationship (QSAR) classification, docking, and molecular dynamics (MD) simulations to investigate the structural basis of selective tau aggregate binding for potential compounds. The QSAR classification model based on the Random Forest algorithm with an accuracy of 96.6% for the selective and 97.6% for the nonselective class of compounds revealed essential selective moieties. The combination of molecular docking, MD simulations, and molecular mechanics Poisson-Boltzmann surface area (MM/PBSA) binding free-energy calculation showed superior binding energy of ligand 63 toward tau and PHF6, a key hexapeptide in tau aggregation, as the most selective compound in the data set. Dissecting the binding properties of ligand 63 and ligand 8 (the least selective compound) within tau and Aβ structures confirmed that these two compounds favor different binding sites of tau; however, the preferential binding site in Aβ was similar for both with lower binding energies calculated for ligand 8. Results revealed that the number of N-heterocycles, the position of nitrogen atoms, and the presence of tertiary amine are important components of selective binding moieties, and they should be maintained in molecules for selective binding to tau aggregates. The predicted structure-selectivity relationship will facilitate the rational design and further development of selective tau imaging agents.
Collapse
Affiliation(s)
- Maryam Gholampour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Hassan Seradj
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Amirhossein Sakhteman
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising 85354, Germany
| |
Collapse
|
6
|
Zhang J, Jin J, Su D, Feng T, Zhao H. Tau-PET imaging in Parkinson's disease: a systematic review and meta-analysis. Front Neurol 2023; 14:1145939. [PMID: 37181568 PMCID: PMC10174250 DOI: 10.3389/fneur.2023.1145939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/30/2023] [Indexed: 05/16/2023] Open
Abstract
Background Pathological tau accumulates in the cerebral cortex of Parkinson's disease (PD), resulting in cognitive deterioration. Positron emission tomography (PET) can be used for in vivo imaging of tau protein. Therefore, we conducted a systematic review and meta-analysis of tau protein burden in PD cognitive impairment (PDCI), PD dementia (PDD), and other neurodegenerative diseases and explored the potential of the tau PET tracer as a biomarker for the diagnosis of PDCI. Methods PubMed, Embase, the Cochrane Library, and Web of Science databases were systematically searched for studies published till 1 June 2022 that used PET imaging to detect tau burden in the brains of PD patients. Standardized mean differences (SMDs) of tau tracer uptake were calculated using random effects models. Subgroup analysis based on the type of tau tracers, meta-regression, and sensitivity analysis was conducted. Results A total of 15 eligible studies were included in the meta-analysis. PDCI patients (n = 109) had a significantly higher tau tracer uptake in the inferior temporal lobe than healthy controls (HCs) (n = 237) and had a higher tau tracer uptake in the entorhinal region than PD with normal cognition (PDNC) patients (n = 61). Compared with progressive supranuclear palsy (PSP) patients (n = 215), PD patients (n = 178) had decreased tau tracer uptake in the midbrain, subthalamic nucleus, globus pallidus, cerebellar deep white matter, thalamus, striatum, substantia nigra, dentate nucleus, red nucleus, putamen, and frontal lobe. Tau tracer uptake values of PD patients (n = 178) were lower than those of patients with Alzheimer's disease (AD) (n = 122) in the frontal lobe and occipital lobe and lower than those in patients with dementia with Lewy bodies (DLB) (n = 55) in the occipital lobe and infratemporal lobe. Conclusion In vivo imaging studies with PET could reveal region-specific binding patterns of the tau tracer in PD patients and help in the differential diagnosis of PD from other neurodegenerative diseases. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/.
Collapse
Affiliation(s)
- Junjiao Zhang
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianing Jin
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dongning Su
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- *Correspondence: Tao Feng
| | - Huiqing Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Huiqing Zhao
| |
Collapse
|
7
|
Rullmann M, Brendel M, Schroeter ML, Saur D, Levin J, Perneczky RG, Tiepolt S, Patt M, Mueller A, Villemagne VL, Classen J, Stephens AW, Sabri O, Barthel H, on behalf of the German Imaging Initiative for Tauopathies (GII4T). Multicenter 18F-PI-2620 PET for In Vivo Braak Staging of Tau Pathology in Alzheimer’s Disease. Biomolecules 2022; 12:biom12030458. [PMID: 35327650 PMCID: PMC8946049 DOI: 10.3390/biom12030458] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 12/04/2022] Open
Abstract
Tau aggregates accumulate in the Alzheimer’s disease (AD) brain according to the established Braak staging scheme and spread from transentorhinal over limbic regions to the neocortex. To impact the management of AD patients, an in vivo tool for tau Braak staging is needed. First-generation tau tracers have limited performance in detecting early stages of tau. Therefore, we tested the corresponding capability of the next-generation tau tracer, 18F-PI-2620. We analyzed 18F-PI-2620 multicenter PET data from 37 beta-amyloid-positive AD dementia patients and those from 26 healthy controls. We applied kinetic modeling of the 0–60 min p.i. PET data using MRTM2 with the lower cerebellum as the reference region to extract Braak stage-dependent distribution volume ratios, whereas controls were used to define Braak stage PET positivity thresholds. Stage-dependent PET positivity widely followed the Braak scheme (except Braak stage III) presenting descending frequency of PET positivity from Braak I (43%), II (38%), III (49%), IV (35%), V (30%) to VI (14%). A strictly hierarchical model was met by 64% of AD dementia cases. Nineteen percent showed a hippocampal sparing tauopathy pattern. Thus, we could assign 87% to the six-stage hierarchical Braak model including tauopathy variants. 18F-PI-2620 PET appears to be able to perform Braak tau staging of AD in vivo.
Collapse
Affiliation(s)
- Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany; (S.T.); (M.P.); (O.S.); (H.B.)
- Correspondence:
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 80336 Munich, Germany;
| | - Matthias L. Schroeter
- Clinic for Cognitive Neurology, University of Leipzig, 04103 Leipzig, Germany;
- Max Planck Institute for Human Cognitive and Brain Sciences, 04103 Leipzig, Germany
| | - Dorothee Saur
- Department of Neurology, University of Leipzig, 04103 Leipzig, Germany; (D.S.); (J.C.)
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany;
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany;
- German Center for Neurodegenerative Disorders (DZNE) Munich, 81377 Munich, Germany
| | - Robert G. Perneczky
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany;
- German Center for Neurodegenerative Disorders (DZNE) Munich, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London W6 8RP, UK
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
| | - Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany; (S.T.); (M.P.); (O.S.); (H.B.)
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany; (S.T.); (M.P.); (O.S.); (H.B.)
| | - Andre Mueller
- Life Molecular Imaging GmbH, 13353 Berlin, Germany; (A.M.); (A.W.S.)
| | | | - Joseph Classen
- Department of Neurology, University of Leipzig, 04103 Leipzig, Germany; (D.S.); (J.C.)
| | | | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany; (S.T.); (M.P.); (O.S.); (H.B.)
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, 04103 Leipzig, Germany; (S.T.); (M.P.); (O.S.); (H.B.)
| | | |
Collapse
|
8
|
Perani D, Cappa SF. The contribution of positron emission tomography to the study of aphasia. HANDBOOK OF CLINICAL NEUROLOGY 2022; 185:151-165. [PMID: 35078596 DOI: 10.1016/b978-0-12-823384-9.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Daniela Perani
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy; In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, Nuclear Medicine Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano F Cappa
- Department of Humanities and Life Sciences, University Institute for Advanced Studies IUSS Pavia, Pavia, Italy; Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy.
| |
Collapse
|
9
|
Xie Y, Wang Y, Jiang S, Xiang X, Wang J, Ning L. Novel strategies for the fight of Alzheimer's disease targeting amyloid-β protein. J Drug Target 2021; 30:259-268. [PMID: 34435898 DOI: 10.1080/1061186x.2021.1973482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD), which is recognised as a devastating neurodegenerative disease throughout the world and lack of effective treatments, is a growing concern in modern society with a growing population of elderly patients. A growing number of studies reveal that abnormal accumulation and deposition of Aβ is responsible for AD. Inspired by this, strategies for the treatment of AD targeting-Aβ clearance have been discussed for a long period, exploring new drugs which is capable of destroying soluble Aβ oligomers and unsolvable Aβ aggregates. In this paper, results of recent clinical trials on several anti-amyloid-β drugs are presented and several emerging anti-amyloid AD therapies based on recent studies are reviewed. Furthermore, some of the current challenges and novel strategies to prevent AD are addressed. Herein, this review focuses on current pharmacotherapy of AD targeting-Aβ and intends to design a promising therapeutic agent for AD treatment.
Collapse
Affiliation(s)
- Yang Xie
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yan Wang
- Chemistry and Chemical Engineering College, Huangshan University, Huangshan, China
| | - Shangfei Jiang
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Xiaohong Xiang
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Jianhua Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, China
| | - Linhong Ning
- Pharmaceutical Engineering Center, Chongqing Medical and Pharmaceutical College, Chongqing, China
| |
Collapse
|
10
|
Abstract
Two pathologically distinct neurodegenerative conditions, progressive supranuclear palsy and corticobasal degeneration, share in common deposits of tau proteins that differ both molecularly and ultrastructurally from the common tau deposits diagnostic of Alzheimer disease. The proteinopathy in these disorders is characterized by fibrillary aggregates of 4R tau proteins. The clinical presentations of progressive supranuclear palsy and of corticobasal degeneration are often confused with more common disorders such as Parkinson disease or subtypes of frontotemporal lobar degeneration. Neither of these 4R tau disorders has effective therapy, and while there are emerging molecular imaging approaches to identify patients earlier in the course of disease, there are as yet no reliably sensitive and specific approaches to diagnoses in life. In this review, aspects of the clinical syndromes, neuropathology, and molecular biomarker imaging studies applicable to progressive supranuclear palsy and to corticobasal degeneration will be presented. Future development of more accurate molecular imaging approaches is proposed.
Collapse
Affiliation(s)
- Kirk A Frey
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, The University of Michigan Health System, Ann Arbor, MI.
| |
Collapse
|
11
|
Oldan JD, Jewells VL, Pieper B, Wong TZ. Complete Evaluation of Dementia: PET and MRI Correlation and Diagnosis for the Neuroradiologist. AJNR Am J Neuroradiol 2021; 42:998-1007. [PMID: 33926896 DOI: 10.3174/ajnr.a7079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 11/14/2020] [Indexed: 12/12/2022]
Abstract
This article will familiarize neuroradiologists with the pathophysiology, clinical findings, and standard MR imaging and PET imaging features of multiple forms of dementia as well as new emerging techniques. Cases were compiled from multiple institutions with the goal of improved diagnostic accuracy and improved patient care as well as information about biomarkers on the horizon. Dementia topics addressed include the following: Alzheimer disease, frontotemporal dementia, cerebral amyloid angiopathy, Lewy body dementia, Parkinson disease and Parkinson disease variants, amyotrophic lateral sclerosis, multisystem atrophy, Huntington disease vascular dementia, and Creutzfeldt-Jakob disease.
Collapse
Affiliation(s)
- J D Oldan
- From the Department of Radiology (J.D.O., V.L.J), University of North Carolina, Chapel Hill, North Carolina
| | - V L Jewells
- From the Department of Radiology (J.D.O., V.L.J), University of North Carolina, Chapel Hill, North Carolina
| | - B Pieper
- Department of Radiology (B.P.), Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - T Z Wong
- Department of Radiology (T.Z.W.), Duke University Hospital, Durham, North Carolina
| |
Collapse
|
12
|
Campese N, Palermo G, Del Gamba C, Beatino MF, Galgani A, Belli E, Del Prete E, Della Vecchia A, Vergallo A, Siciliano G, Ceravolo R, Hampel H, Baldacci F. Progress regarding the context-of-use of tau as biomarker of Alzheimer's disease and other neurodegenerative diseases. Expert Rev Proteomics 2021; 18:27-48. [PMID: 33545008 DOI: 10.1080/14789450.2021.1886929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Tau protein misfolding and accumulation in toxic species is a critical pathophysiological process of Alzheimer's disease (AD) and other neurodegenerative disorders (NDDs). Tau biomarkers, namely cerebrospinal fluid (CSF) total-tau (t-tau), 181-phosphorylated tau (p-tau), and tau-PET tracers, have been recently embedded in the diagnostic criteria for AD. Nevertheless, the role of tau as a diagnostic and prognostic biomarker for other NDDs remains controversial.Areas covered: We performed a systematical PubMed-based review of the most recent advances in tau-related biomarkers for NDDs. We focused on papers published from 2015 to 2020 assessing the diagnostic or prognostic value of each biomarker.Expert opinion: The assessment of tau biomarkers in alternative easily accessible matrices, through the development of ultrasensitive techniques, represents the most significant perspective for AD-biomarker research. In NDDs, novel tau isoforms (e.g. p-tau217) or proteolytic fragments (e.g. N-terminal fragments) may represent candidate diagnostic and prognostic biomarkers and may help monitoring disease progression. Protein misfolding amplification assays, allowing the identification of different tau strains (e.g. 3 R- vs. 4 R-tau) in CSF, may constitute a breakthrough for the in vivo stratification of NDDs. Tau-PET may help tracking the spatial-temporal evolution of tau pathophysiology in AD but its application outside the AD-spectrum deserves further studies.
Collapse
Affiliation(s)
- Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Palermo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudia Del Gamba
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Del Prete
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Andrea Vergallo
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| |
Collapse
|
13
|
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: 105] [Impact Index Per Article: 26.3] [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
|
14
|
Abstract
The history of Alzheimer's disease (AD) started in 1907, but we needed to wait until the end of the century to identify the components of pathological hallmarks and genetic subtypes and to formulate the first pathogenic hypothesis. Thanks to biomarkers and new technologies, the concept of AD then rapidly changed from a static view of an amnestic dementia of the presenium to a biological entity that could be clinically manifested as normal cognition or dementia of different types. What is clearly emerging from studies is that AD is heterogeneous in each aspect, such as amyloid composition, tau distribution, relation between amyloid and tau, clinical symptoms, and genetic background, and thus it is probably impossible to explain AD with a single pathological process. The scientific approach to AD suffers from chronological mismatches between clinical, pathological, and technological data, causing difficulty in conceiving diagnostic gold standards and in creating models for drug discovery and screening. A recent mathematical computer-based approach offers the opportunity to study AD in real life and to provide a new point of view and the final missing pieces of the AD puzzle.
Collapse
Affiliation(s)
- Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
15
|
Drzezga A, Bischof GN, Giehl K, van Eimeren T. PET and SPECT Imaging of Neurodegenerative Diseases. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00085-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
16
|
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: 22] [Impact Index Per Article: 4.4] [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
|
17
|
Charil A, Shcherbinin S, Southekal S, Devous MD, Mintun M, Murray ME, Miller BB, Schwarz AJ. Tau Subtypes of Alzheimer's Disease Determined in vivo Using Flortaucipir PET Imaging. J Alzheimers Dis 2020; 71:1037-1048. [PMID: 31476153 DOI: 10.3233/jad-190264] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
At autopsy, individuals with Alzheimer's disease (AD) exhibit heterogeneity in the distribution of neurofibrillary tangles in neocortical and hippocampal regions. Subtypes of AD, defined using an algorithm based on the relative number of tangle counts in these regions, have been proposed-hippocampal sparing (relative sparing of the hippocampus but high cortical load), limbic predominant (high hippocampal load but lower load in association cortices), and typical (balanced neurofibrillary tangles counts in the hippocampus and association cortices) AD-and shown to be associated with distinct antemortem clinical phenotypes. The ability to distinguish these AD subtypes from the more typical tau signature in vivo could have important implications for clinical research. Flortaucipir positron emission tomography (PET) images acquired from 45 amyloid-positive participants, defined clinically as mild cognitive impairment or AD, aged 50-92 years, 56% female, and estimated to be Braak V-VI based on their PET pattern of tau pathology, were studied. By translating the neuropathologic algorithm to flortaucipir PET scans, patterns of tau pathology consistent with autopsy findings, and with a similar prevalence, were identified in vivo. 6/45 (13%) participants were identified as hippocampal sparing and 6/45 (13%) as limbic predominant AD subtypes. Hippocampal sparing participants were significantly younger than those assigned to the other two subtypes. Worse performance on delayed recall was associated with increased hippocampal tau signal, and worse performance on the trail making test B-A was associated with lower values of the hippocampus to cortex ratio. Prospective studies can further validate the flortaucipir SUVR cut-points and the phenotype of the corresponding AD subtypes.
Collapse
Affiliation(s)
| | | | | | | | - Mark Mintun
- Eli Lilly and Company, Indianapolis, IN, USA.,Avid Radiopharmaceuticals, Philadelphia, PA, USA
| | | | | | - Adam J Schwarz
- Eli Lilly and Company, Indianapolis, IN, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA.,Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
18
|
A critical review of radiotracers in the positron emission tomography imaging of traumatic brain injury: FDG, tau, and amyloid imaging in mild traumatic brain injury and chronic traumatic encephalopathy. Eur J Nucl Med Mol Imaging 2020; 48:623-641. [DOI: 10.1007/s00259-020-04926-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
|
19
|
Early-phase [ 18F]PI-2620 tau-PET imaging as a surrogate marker of neuronal injury. Eur J Nucl Med Mol Imaging 2020; 47:2911-2922. [PMID: 32318783 PMCID: PMC7567714 DOI: 10.1007/s00259-020-04788-w] [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: 01/02/2020] [Accepted: 03/24/2020] [Indexed: 12/31/2022]
Abstract
Purpose Second-generation tau radiotracers for use with positron emission tomography (PET) have been developed for visualization of tau deposits in vivo. For several β-amyloid and first-generation tau-PET radiotracers, it has been shown that early-phase images can be used as a surrogate of neuronal injury. Therefore, we investigated the performance of early acquisitions of the novel tau-PET radiotracer [18F]PI-2620 as a potential substitute for [18F]fluorodeoxyglucose ([18F]FDG). Methods Twenty-six subjects were referred with suspected tauopathies or overlapping parkinsonian syndromes (Alzheimer’s disease, progressive supranuclear palsy, corticobasal syndrome, multi-system atrophy, Parkinson’s disease, multi-system atrophy, Parkinson's disease, frontotemporal dementia) and received a dynamic [18F]PI-2620 tau-PET (0–60 min p.i.) and static [18F]FDG-PET (30–50 min p.i.). Regional standardized uptake value ratios of early-phase images (single frame SUVr) and the blood flow estimate (R1) of [18F]PI-2620-PET were correlated with corresponding quantification of [18F]FDG-PET (global mean/cerebellar normalization). Reduced tracer uptake in cortical target regions was also interpreted visually using 3-dimensional stereotactic surface projections by three more and three less experienced readers. Spearman rank correlation coefficients were calculated between early-phase [18F]PI-2620 tau-PET and [18F]FDG-PET images for all cortical regions and frequencies of disagreement between images were compared for both more and less experienced readers. Results Highest agreement with [18F]FDG-PET quantification was reached for [18F]PI-2620-PET acquisition from 0.5 to 2.5 min p.i. for global mean (lowest R = 0.69) and cerebellar scaling (lowest R = 0.63). Correlation coefficients (summed 0.5–2.5 min SUVr & R1) displayed strong agreement in all cortical target regions for global mean (RSUVr 0.76, RR1 = 0.77) and cerebellar normalization (RSUVr 0.68, RR1 = 0.68). Visual interpretation revealed high regional correlations between early-phase tau-PET and [18F]FDG-PET. There were no relevant differences between more and less experienced readers. Conclusion Early-phase imaging of [18F]PI-2620 can serve as a surrogate biomarker for neuronal injury. Dynamic imaging or a dual time-point protocol for tau-PET imaging could supersede additional [18F]FDG-PET imaging by indexing both the distribution of tau and the extent of neuronal injury. Electronic supplementary material The online version of this article (10.1007/s00259-020-04788-w) contains supplementary material, which is available to authorized users.
Collapse
|
20
|
Kuang G, Murugan NA, Zhou Y, Nordberg A, Ågren H. Computational Insight into the Binding Profile of the Second-Generation PET Tracer PI2620 with Tau Fibrils. ACS Chem Neurosci 2020; 11:900-908. [PMID: 32069017 DOI: 10.1021/acschemneuro.9b00578] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abnormal deposition of hyperphosphorylated tau as neurofibrillary tangles (NFTs) is an important pathological hallmark of Alzheimer's disease (AD) and of other neurodegenerative disorders. A noninvasive positron emission tomography (PET) tracer that quantifies neurofibrillary tangles in vivo can enhance the clinical diagnosis of AD and can also be used to evaluate the efficacy of therapeutics aimed at reducing the abnormal aggregation of the tau fibril in the brain. In this paper, we study the binding profile of fibrillar tau aggregates with a PET tracer PI2620, which is a new second generation tau PET tracer that is presently experimentally and clinically studied. The target structure for the tau fibril is based on cryo-electron microscopy (cryo-EM) structure. A multiscale simulation workflow including molecular docking, molecular dynamics simulation, metadynamics simulation, and free energy calculations was implemented. We find that PI2620 can bind to eight surface binding sites, three core binding sites, and one entry site. The binding at the core sites and entry site is found to be much more favorable than that on the surface sites due to stronger hydrophobic interactions and less solvent exposure. Furthermore, the entry site which is formed by the terminal β-sheets of the fibril is found to have the highest binding affinity to PI2620. Importantly, the binding capacity at the entry site can be much higher than that at other core sites, due to its easy accessibility. Therefore, the entry site is believed to be the major binding site for PI2620. A previous computational study on tracers with tau fibrils reports a maximum of four binding sites. Through use of methods that allow us to locate "cryptic binding sites", we report here additional core sites available for binding and we address the limitation of using the cryo-EM structure alone for structure-based tracer design. Our results could be helpful for elucidating the binding mechanism of imaging tracers with the fibrillar form of tau, a knowledge that in turn can be used to guide the development of compounds with higher affinity and selectivity for tau using structure-based design strategies.
Collapse
Affiliation(s)
- Guanglin Kuang
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, Stockholm 10691, Sweden
| | - N. Arul Murugan
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, Stockholm 10691, Sweden
| | - Yang Zhou
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, Stockholm 10691, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Center for Alzheimer Research, Stockholm 17177, Sweden
- Aging Theme, Karolinska University Hospital, Stockholm 14186, Sweden
| | - Hans Ågren
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, Stockholm 10691, Sweden
- College of Chemistry and Chemical Engineering, Henan University, Kaifeng, Henan 475004, P. R. China
| |
Collapse
|
21
|
Uzuegbunam BC, Librizzi D, Hooshyar Yousefi B. PET Radiopharmaceuticals for Alzheimer's Disease and Parkinson's Disease Diagnosis, the Current and Future Landscape. Molecules 2020; 25:E977. [PMID: 32098280 PMCID: PMC7070523 DOI: 10.3390/molecules25040977] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
Ironically, population aging which is considered a public health success has been accompanied by a myriad of new health challenges, which include neurodegenerative disorders (NDDs), the incidence of which increases proportionally to age. Among them, Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common, with the misfolding and the aggregation of proteins being common and causal in the pathogenesis of both diseases. AD is characterized by the presence of hyperphosphorylated τ protein (tau), which is the main component of neurofibrillary tangles (NFTs), and senile plaques the main component of which is β-amyloid peptide aggregates (Aβ). The neuropathological hallmark of PD is α-synuclein aggregates (α-syn), which are present as insoluble fibrils, the primary structural component of Lewy body (LB) and neurites (LN). An increasing number of non-invasive PET examinations have been used for AD, to monitor the pathological progress (hallmarks) of disease. Notwithstanding, still the need for the development of novel detection tools for other proteinopathies still remains. This review, although not exhaustively, looks at the timeline of the development of existing tracers used in the imaging of Aβ and important moments that led to the development of these tracers.
Collapse
Affiliation(s)
| | - Damiano Librizzi
- Department of Nuclear Medicine, Philipps-University of Marburg, 35043 Marburg, Germany;
| | - Behrooz Hooshyar Yousefi
- Nuclear Medicine Department, and Neuroimaging Center, Technical University of Munich, 81675 Munich, Germany;
- Department of Nuclear Medicine, Philipps-University of Marburg, 35043 Marburg, Germany;
| |
Collapse
|
22
|
Al-Janabi OM, Brown CA, Bahrani AA, Abner EL, Barber JM, Gold BT, Goldstein LB, Murphy RR, Nelson PT, Johnson NF, Shaw LM, Smith CD, Trojanowski JQ, Wilcock DM, Jicha GA. Distinct White Matter Changes Associated with Cerebrospinal Fluid Amyloid-β1-42 and Hypertension. J Alzheimers Dis 2019; 66:1095-1104. [PMID: 30400099 DOI: 10.3233/jad-180663] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) pathology and hypertension (HTN) are risk factors for development of white matter (WM) alterations and might be independently associated with these alterations in older adults. OBJECTIVE To evaluate the independent and synergistic effects of HTN and AD pathology on WM alterations. METHODS Clinical measures of cerebrovascular disease risk were collected from 62 participants in University of Kentucky Alzheimer's Disease Center studies who also had cerebrospinal fluid (CSF) sampling and MRI brain scans. CSF Aβ1-42 levels were measured as a marker of AD, and fluid-attenuated inversion recovery imaging and diffusion tensor imaging were obtained to assess WM macro- and microstructural properties. Linear regression analyses were used to assess the relationships among WM alterations, cerebrovascular disease risk, and AD pathology. Voxelwise analyses were performed to examine spatial patterns of WM alteration associated with each pathology. RESULTS HTN and CSF Aβ1-42 levels were each associated with white matter hyperintensities (WMH). Also, CSF Aβ1-42 levels were associated with alterations in normal appearing white matter fractional anisotropy (NAWM-FA), whereas HTN was marginally associated with alterations in NAWM-FA. Linear regression analyses demonstrated significant main effects of HTN and CSF Aβ1-42 on WMH volume, but no significant HTN×CSF Aβ1-42 interaction. Furthermore, voxelwise analyses showed unique patterns of WM alteration associated with hypertension and CSF Aβ1-42. CONCLUSION Associations of HTN and lower CSF Aβ1-42 with WM alteration were statistically and spatially distinct, suggesting independent rather than synergistic effects. Considering such spatial distributions may improve diagnostic accuracy to address each underlying pathology.
Collapse
Affiliation(s)
- Omar M Al-Janabi
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Behavioral Science, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Christopher A Brown
- Departments of Neuroscience, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Ahmed A Bahrani
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Biomedical Engineering, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Epidemiology and Biostatistics, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Justin M Barber
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Brian T Gold
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Neuroscience, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Larry B Goldstein
- Departments of Neurology, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Ronan R Murphy
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Neurology, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Pathology, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Nathan F Johnson
- Departments of Rehabilitation Science, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Leslie M Shaw
- Department of Pathology & Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Charles D Smith
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Neurology, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - John Q Trojanowski
- Department of Pathology & Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Physiology, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Behavioral Science, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA.,Departments of Neurology, University of Kentucky Colleges of Medicine, Public Health, Health Sciences and Engineering Lexington, KY, USA
| |
Collapse
|
23
|
Mainta IC, Vargas MI, Trombella S, Frisoni GB, Unschuld PG, Garibotto V. Hybrid PET-MRI in Alzheimer's Disease Research. Methods Mol Biol 2019; 1750:185-200. [PMID: 29512073 DOI: 10.1007/978-1-4939-7704-8_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Multiple factors, namely amyloid, tau, inflammation, metabolic, and perfusion changes, contribute to the cascade of neurodegeneration and functional decline occurring in Alzheimer's disease (AD). These molecular and cellular processes and related functional and morphological changes can be visualized in vivo by two imaging modalities, namely positron emission tomography (PET) and magnetic resonance imaging (MRI). These imaging biomarkers are now part of the diagnostic algorithm and of particular interest for patient stratification and targeted drug development.In this field the availability of hybrid PET/MR systems not only offers a comprehensive evaluation in a single imaging session, but also opens new possibilities for the integration of the two imaging information. Here, we cover the clinical protocols and practical details of FDG, amyloid, and tau PET/MR imaging as applied in our institutions.
Collapse
Affiliation(s)
- Ismini C Mainta
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland. .,Faculty of Medicine, Nuclear Medicine Department, Geneva University Medical Center, University of Geneva, Geneva, Switzerland.
| | - Maria I Vargas
- Faculty of Medicine, Nuclear Medicine Department, Geneva University Medical Center, University of Geneva, Geneva, Switzerland.,Division of Neuroradiology, Geneva University Hospitals, Geneva, Switzerland
| | - Sara Trombella
- Faculty of Medicine, Nuclear Medicine Department, Geneva University Medical Center, University of Geneva, Geneva, Switzerland
| | - Giovanni B Frisoni
- Faculty of Medicine, Nuclear Medicine Department, Geneva University Medical Center, University of Geneva, Geneva, Switzerland.,Department of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Paul G Unschuld
- Institute for Regenerative Medicine and Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Nuclear Medicine Department, Geneva University Medical Center, University of Geneva, Geneva, Switzerland
| |
Collapse
|
24
|
Tan CC, Zhang XY, Tan L, Yu JT. Tauopathies: Mechanisms and Therapeutic Strategies. J Alzheimers Dis 2019; 61:487-508. [PMID: 29278892 DOI: 10.3233/jad-170187] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tauopathies are morphologically, biochemically, and clinically heterogeneous neurodegenerative diseases defined by the accumulation of abnormal tau proteins in the brain. There is no effective method to prevent and reverse the tauopathies, but this gloomy picture has been changed by recent research advances. Evidences from genetic studies, experimental animal models, and molecular and cell biology have shed light on the main mechanisms of the diseases. The development of radiology and biochemistry, especially the development of PET imaging, will provide important biomarkers for the clinical diagnosis and treatment. Given the central role of tau in tauopathies, many treatments have constantly emerged, including targeting phosphorylation, targeting aggregation, increasing microtubule stabilization, tau immunization, clearance of tau, anti-inflammatory treatment, and other therapeutics. There is still a long way to go before we obtain drug therapy targeted at multifactor mechanisms.
Collapse
Affiliation(s)
- Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xiao-Yan Zhang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
25
|
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: 426] [Impact Index Per Article: 71.0] [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
|
26
|
Snyder HM, Carare RO, DeKosky ST, de Leon MJ, Dykxhoorn D, Gan L, Gardner R, Hinds SR, Jaffee M, Lamb BT, Landau S, Manley G, McKee A, Perl D, Schneider JA, Weiner M, Wellington C, Yaffe K, Bain L, Pacifico AM, Carrillo MC. Military-related risk factors for dementia. Alzheimers Dement 2018; 14:1651-1662. [PMID: 30415806 PMCID: PMC6281800 DOI: 10.1016/j.jalz.2018.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION In recent years, there has been growing discussion to better understand the pathophysiological mechanisms of traumatic brain injury and post-traumatic stress disorder and how they may be linked to an increased risk of neurodegenerative diseases including Alzheimer's disease in veterans. METHODS Building on that discussion, and subsequent to a special issue of Alzheimer's & Dementia published in June 2014, which focused on military risk factors, the Alzheimer's Association convened a continued discussion of the scientific community on December 1, 2016. RESULTS During this meeting, participants presented and evaluated progress made since 2012 and identified outstanding knowledge gaps regarding factors that may impact veterans' risk for later life dementia. DISCUSSION The following is a summary of the invited presentations and moderated discussions of both the review of scientific understanding and identification of gaps to inform further investigations.
Collapse
Affiliation(s)
- Heather M Snyder
- Medical & Scientific Relations, Alzheimer's Association, Chicago, IL, USA.
| | - Roxana O Carare
- Clinical Neuroanatomy, Equality and Diversity Lead, University of Southampton, Southampton, United Kingdom
| | - Steven T DeKosky
- Department of Neurology and Neuroscience, University of Florida, Gainesville, FL, USA
| | - Mony J de Leon
- Department of Psychiatry, New York University Medical Center, New York City, NY, USA
| | - Derek Dykxhoorn
- Department of Microbiology and Immunology, Miami University, Miami, FL, USA
| | - Li Gan
- Gladstone Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Raquel Gardner
- Department of Psychiatry, Neurology & Epidemiology, University of California, San Francisco, San Francisco, CA, USA
| | - Sidney R Hinds
- Blast Injury Research Program Coordinating Office, United States Army Medical Research and Material Command, Frederick, MD, USA
| | - Michael Jaffee
- Department of Neurology and Neuroscience, University of Florida, Gainesville, FL, USA
| | - Bruce T Lamb
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA
| | - Susan Landau
- Helen Willis Neuroscience Institute, University of California, Berkley, Berkley, CA, USA
| | - Geoff Manley
- Department of Psychiatry, Neurology & Epidemiology, University of California, San Francisco, San Francisco, CA, USA
| | - Ann McKee
- Department of Neurology and Pathology, Boston University, Boston, MA, USA
| | - Daniel Perl
- Department of Pathology, Uniformed Services University, Bethesda, MD, USA
| | - Julie A Schneider
- Neurology Department, Rush University Medical Center, Chicago, IL, USA
| | - Michael Weiner
- Department of Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Cheryl Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kristine Yaffe
- Department of Psychiatry, Neurology & Epidemiology, University of California, San Francisco, San Francisco, CA, USA
| | - Lisa Bain
- Independent Science Writer, Philadelphia, PA, USA
| | | | - Maria C Carrillo
- Medical & Scientific Relations, Alzheimer's Association, Chicago, IL, USA
| |
Collapse
|
27
|
Tau PET imaging evidence in patients with cognitive impairment: preparing for clinical use. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0297-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
28
|
A Closer Look into the Role of Protein Tau in the Identification of Promising Therapeutic Targets for Alzheimer's Disease. Brain Sci 2018; 8:brainsci8090162. [PMID: 30149687 PMCID: PMC6162660 DOI: 10.3390/brainsci8090162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 01/09/2023] Open
Abstract
One of the most commonly known chronic neurodegenerative disorders, Alzheimer's disease (AD), manifests the common type of dementia in 60⁻80% of cases. From a clinical standpoint, a patent cognitive decline and a severe change in personality, as caused by a loss of neurons, is usually evident in AD with about 50 million people affected in 2016. The disease progression in patients is distinguished by a gradual plummet in cognitive functions, eliciting symptoms such as memory loss, and eventually requiring full-time medical care. From a histopathological standpoint, the defining characteristics are intracellular aggregations of hyper-phosphorylated tau protein, known as neurofibrillary tangles (NFT), and depositions of amyloid β-peptides (Aβ) in the brain. The abnormal phosphorylation of tau protein is attributed to a wide gamut of neurological disorders known as tauopathies. In addition to the hyperphosphorylated tau lesions, neuroinflammatory processes could occur in a sustained manner through astro-glial activation, resulting in the disease progression. Recent findings have suggested a strong interplay between the mechanism of Tau phosphorylation, disruption of microtubules, and synaptic loss and pathology of AD. The mechanisms underlying these interactions along with their respective consequences in Tau pathology are still ill-defined. Thus, in this review: (1) we highlight the interplays existing between Tau pathology and AD; and (2) take a closer look into its role while identifying some promising therapeutic advances including state of the art imaging techniques.
Collapse
|
29
|
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: 92] [Impact Index Per Article: 13.1] [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
|
30
|
Rafique W, Kramer V, Pardo T, Smits R, Spilhaug MM, Hoepping A, Savio E, Engler H, Kuljs R, Amaral H, Riss PJ. Image-Guided Development of Heterocyclic Sulfoxides as Ligands for Tau Neurofibrillary Tangles: From First-in-Man to Second-Generation Ligands. ACS OMEGA 2018; 3:7567-7579. [PMID: 30087917 PMCID: PMC6068598 DOI: 10.1021/acsomega.8b00975] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/15/2018] [Indexed: 06/08/2023]
Abstract
Positron emission tomography (PET) imaging of misfolded protein aggregates that form in neurodegenerative processes of the brain is key to providing a robust marker for improved diagnosis and evaluation of treatments. We report the development of advanced radiotracer candidates based on the sulfoxide scaffold found in proton pump inhibitors (lansoprazole, prevacid) with inherent affinity to neurofibrillary tangles in Alzheimer's disease and related disorders (e.g., dementia with Lewy bodies and the frontotemporal degeneration syndrome). First-in-man results obtained with [18F]lansoprazole and N-methyl-[18F]lansoprazole were used to guide the design of a set of 24 novel molecules with suitable properties for neuroimaging with PET. Compounds were synthesized and characterized pharmacologically, and the binding affinity of the compounds to synthetic human tau-441 fibrils was determined. Selectivity of binding was assessed using α-synuclein and β-amyloid fibrils to address the key misfolded proteins of relevance in dementia. To complete the pharmacokinetic profiling in vitro, plasma protein binding and lipophilicity were investigated. Highly potent and selective new radiotracer candidates were identified for further study.
Collapse
Affiliation(s)
- Waqas Rafique
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands
vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Vasko Kramer
- Positronpharma
SA, Rancagua 878, 7500921 Providencia, Santiago, Chile
- Center
of Nuclear Medicine Positronmed, Julio Prado 714, 7501068 Providencia, Santiago, Chile
| | - Tania Pardo
- Departamento
de Montevideo, Uruguayan Centre of Molecular
Imaging (CUDIM), Av.
Dr. Américo Ricaldoni 2010, 11600 Montevideo, Uruguay
| | - René Smits
- Advanced
Biochemical Compounds GmbH, Heinrich-Glaeser-Strasse 10-14, D-01454 Radeberg, Germany
| | - Mona M. Spilhaug
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands
vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Alexander Hoepping
- Advanced
Biochemical Compounds GmbH, Heinrich-Glaeser-Strasse 10-14, D-01454 Radeberg, Germany
| | - Eduardo Savio
- Departamento
de Montevideo, Uruguayan Centre of Molecular
Imaging (CUDIM), Av.
Dr. Américo Ricaldoni 2010, 11600 Montevideo, Uruguay
| | - Henry Engler
- Departamento
de Montevideo, Uruguayan Centre of Molecular
Imaging (CUDIM), Av.
Dr. Américo Ricaldoni 2010, 11600 Montevideo, Uruguay
| | - Rodrigo Kuljs
- Zdrav
Mozak Clinical Neuroscience Center, Julio Prado 714, 7501068 Providencia, Santiago, Chile
| | - Horacio Amaral
- Positronpharma
SA, Rancagua 878, 7500921 Providencia, Santiago, Chile
- Center
of Nuclear Medicine Positronmed, Julio Prado 714, 7501068 Providencia, Santiago, Chile
| | - Patrick J. Riss
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands
vei 26, Kjemibygningen, 0371 Oslo, Norway
- Klinik
for Kirurgi og Nevrofag, Oslo Universitets
Sykehus HF—Rikshospitalet, Postboks
4950 Nydalen, 0424 Oslo, Norway
- Norsk
Medisinsk Syklotronsenter AS, Gaustad,
Postboks 4950 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
31
|
Murugan NA, Nordberg A, Ågren H. Different Positron Emission Tomography Tau Tracers Bind to Multiple Binding Sites on the Tau Fibril: Insight from Computational Modeling. ACS Chem Neurosci 2018; 9:1757-1767. [PMID: 29630333 DOI: 10.1021/acschemneuro.8b00093] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Using the recently reported cryo-EM structure for the tau fibril [ Fitzpatrick et al. (2017) Nature 547, 185-190 ], which is a potential target concerning Alzheimer's disease, we present the first molecular modeling studies on its interaction with various positron emission tomography (PET) tracers. Experimentally, based on the binding assay studies, at least three different high-affinity binding sites have been reported for tracers in the tau fibril. Herein, through integrated modeling using molecular docking, molecular dynamics, and binding free energy calculations, we provide insight into the binding patterns of various tracers to the tau fibril. We suggest that there are four different high-affinity binding sites available for many of the studied tracers showing varying binding affinity to different binding sites. Thus, PBB3 binds most strongly to site 4, and interestingly, this site is not a preferable site for any other tracers. For THK5351, our data show that it strongly binds to sites 3 and 1, the former one being more preferable. We also find that MK6240 and T807 bind to site 1 specifically. The modeling data also give some insight into whether a tracer bound to a specific site can be replaced by others or not. For example, the displacement of T807 by PBB3 as reported experimentally can also be explained and attributed to the larger binding affinity of the latter compound in all binding sites. The binding free energy results explain very well the small binding affinity of THK523 compared to all the aryl quinoline moieties containing THK tracers. The ability of certain tau tracers, like FDDNP and THK523, to bind to amyloid fibrils has also been investigated. Furthermore, such off-target interaction of tau tracers with amyloid beta fibrils has been validated using a quantum mechanical fragmentation approach.
Collapse
Affiliation(s)
- N. Arul Murugan
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center of Alzheimer Research, Division of Clinical Geriatric, Karolinska Institutet, Huddinge, S-141 86 Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, S-141 86 Stockholm, Sweden
| | - Hans Ågren
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
- Department of Physics and Astronomy, Uppsala University, SE-751 20 Uppsala, Sweden
| |
Collapse
|
32
|
Aldag M, Armstrong RC, Bandak F, Bellgowan PSF, Bentley T, Biggerstaff S, Caravelli K, Cmarik J, Crowder A, DeGraba TJ, Dittmer TA, Ellenbogen RG, Greene C, Gupta RK, Hicks R, Hoffman S, Latta RC, Leggieri MJ, Marion D, Mazzoli R, McCrea M, O'Donnell J, Packer M, Petro JB, Rasmussen TE, Sammons-Jackson W, Shoge R, Tepe V, Tremaine LA, Zheng J. The Biological Basis of Chronic Traumatic Encephalopathy following Blast Injury: A Literature Review. J Neurotrauma 2018; 34:S26-S43. [PMID: 28937953 DOI: 10.1089/neu.2017.5218] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The United States Department of Defense Blast Injury Research Program Coordinating Office organized the 2015 International State-of-the-Science meeting to explore links between blast-related head injury and the development of chronic traumatic encephalopathy (CTE). Before the meeting, the planning committee examined articles published between 2005 and October 2015 and prepared this literature review, which summarized broadly CTE research and addressed questions about the pathophysiological basis of CTE and its relationship to blast- and nonblast-related head injury. It served to inform participants objectively and help focus meeting discussion on identifying knowledge gaps and priority research areas. CTE is described generally as a progressive neurodegenerative disorder affecting persons exposed to head injury. Affected individuals have been participants primarily in contact sports and military personnel, some of whom were exposed to blast. The symptomatology of CTE overlaps with Alzheimer's disease and includes neurological and cognitive deficits, psychiatric and behavioral problems, and dementia. There are no validated diagnostic criteria, and neuropathological evidence of CTE has come exclusively from autopsy examination of subjects with histories of exposure to head injury. The perivascular accumulation of hyperphosphorylated tau (p-tau) at the depths of cortical sulci is thought to be unique to CTE and has been proposed as a diagnostic requirement, although the contribution of p-tau and other reported pathologies to the development of clinical symptoms of CTE are unknown. The literature on CTE is limited and is focused predominantly on head injuries unrelated to blast exposure (e.g., football players and boxers). In addition, comparative analyses of clinical case reports has been challenging because of small case numbers, selection biases, methodological differences, and lack of matched controls, particularly for blast-exposed individuals. Consequently, the existing literature is not sufficient to determine whether the development of CTE is associated with head injury frequency (e.g., single vs. multiple exposures) or head injury type (e.g., impact, nonimpact, blast-related). Moreover, the incidence and prevalence of CTE in at-risk populations is unknown. Future research priorities should include identifying additional risk factors, pursuing population-based longitudinal studies, and developing the ability to detect and diagnose CTE in living persons using validated criteria.
Collapse
Affiliation(s)
- Matt Aldag
- 1 Booz Allen Hamilton , McLean, Virginia
| | - Regina C Armstrong
- 2 Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Faris Bandak
- 3 Defense Advanced Research Projects Agency , Arlington, Virginia
| | | | | | - Sean Biggerstaff
- 6 Office of the Assistant Secretary of Defense , Health Affairs, Falls Church, Virginia
| | | | - Joan Cmarik
- 7 Office of the Principal Assistant for Acquisition, United States Army Medical Research and Materiel Command , Frederick, Maryland
| | - Alicia Crowder
- 8 Combat Casualty Care Research Program , United States Army Medical Research and Materiel Command, Fort Detrick, Maryland
| | | | | | - Richard G Ellenbogen
- 10 Departments of Neurological Surgery and Global Health Medicine, University of Washington , Seattle, Washington
| | - Colin Greene
- 11 Joint Trauma Analysis and Prevention of Injuries in Combat Program, Frederick, Maryland
| | - Raj K Gupta
- 12 Department of Defense Blast Injury Research Program Coordinating Office, United States Army Medical Research and Materiel Command , Frederick, Maryland
| | | | | | | | - Michael J Leggieri
- 12 Department of Defense Blast Injury Research Program Coordinating Office, United States Army Medical Research and Materiel Command , Frederick, Maryland
| | - Donald Marion
- 16 Defense and Veterans Brain Injury Center , Silver Spring, Maryland
| | | | | | | | - Mark Packer
- 20 Hearing Center of Excellence , Lackland, Texas
| | - James B Petro
- 21 Office of the Assistant Secretary of Defense, Research and Engineering, Arlington, Virginia
| | - Todd E Rasmussen
- 8 Combat Casualty Care Research Program , United States Army Medical Research and Materiel Command, Fort Detrick, Maryland
| | - Wendy Sammons-Jackson
- 22 Office of the Principal Assistant for Research and Technology , United States Army Medical Research and Materiel Command, Fort Detrick, Maryland
| | - Richard Shoge
- 23 Military Operational Medicine Research Program, United States Army Medical Research and Materiel Command , Fort Detrick, Maryland
| | | | | | - James Zheng
- 25 Program Executive Office Soldier , Fort Belvoir, Virginia
| |
Collapse
|
33
|
Susceptibility of brain atrophy to TRIB3 in Alzheimer's disease, evidence from functional prioritization in imaging genetics. Proc Natl Acad Sci U S A 2018; 115:3162-3167. [PMID: 29511103 DOI: 10.1073/pnas.1706100115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The joint modeling of brain imaging information and genetic data is a promising research avenue to highlight the functional role of genes in determining the pathophysiological mechanisms of Alzheimer's disease (AD). However, since genome-wide association (GWA) studies are essentially limited to the exploration of statistical correlations between genetic variants and phenotype, the validation and interpretation of the findings are usually nontrivial and prone to false positives. To address this issue, in this work, we investigate the functional genetic mechanisms underlying brain atrophy in AD by studying the involvement of candidate variants in known genetic regulatory functions. This approach, here termed functional prioritization, aims at testing the sets of gene variants identified by high-dimensional multivariate statistical modeling with respect to known biological processes to introduce a biology-driven validation scheme. When applied to the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort, the functional prioritization allowed for identifying a link between tribbles pseudokinase 3 (TRIB3) and the stereotypical pattern of gray matter loss in AD, which was confirmed in an independent validation sample, and that provides evidence about the relation between this gene and known mechanisms of neurodegeneration.
Collapse
|
34
|
Zhu B, Zhang T, Jiang Q, Li Y, Fu Y, Dai J, Li G, Qi Q, Cheng Y. Synthesis and evaluation of pyrazine and quinoxaline fluorophores for in vivo detection of cerebral tau tangles in Alzheimer's models. Chem Commun (Camb) 2018; 54:11558-11561. [PMID: 30204164 DOI: 10.1039/c8cc06897f] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The quinoxaline derivative 3b is a candidate probe for fluorescence turn-on detection of tau tangles both in vitro and in mice in vivo.
Collapse
Affiliation(s)
- Biyue Zhu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Ting Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Qian Jiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Ying Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Yu Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering
- South-Central University for Nationalities
- Wuhan 430074
- China
| | - Ge Li
- Guangdong Laboratory Animals Monitoring Institute
- Guangdong Provincial Key Laboratory of Laboratory Animals
- Guangzhou 510663
- China
| | - Qingrong Qi
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Yan Cheng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry
- Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology
- West China School of Pharmacy
- Sichuan University
- Chengdu
| |
Collapse
|
35
|
Devous MD, Joshi AD, Navitsky M, Southekal S, Pontecorvo MJ, Shen H, Lu M, Shankle WR, Seibyl JP, Marek K, Mintun MA. Test-Retest Reproducibility for the Tau PET Imaging Agent Flortaucipir F 18. J Nucl Med 2017; 59:937-943. [PMID: 29284675 DOI: 10.2967/jnumed.117.200691] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/08/2017] [Indexed: 01/16/2023] Open
Abstract
Alzheimer disease (AD) is characterized by β-amyloid (Aβ) plaques and tau neurofibrillary tangles. There are several PET imaging biomarkers for Aβ including 11C-PiB and 18F-florbetapir. Recently, PET tracers for tau neurofibrillary tangles have become available and have shown utility in detection and monitoring of neurofibrillary pathology over time. Flortaucipir F 18 is one such tracer. Initial clinical studies indicated greater tau binding in AD and mild cognitive impairment patients than in controls in a pattern consistent with tau pathology observed at autopsy. However, little is known about the reproducibility of such findings. To our knowledge, this study reports the first data regarding test-retest reproducibility of flortaucipir F 18 PET. Methods: Twenty-one subjects who completed the study (5 healthy controls, 6 mild cognitive impairment, and 10 AD) received 370 MBq of flortaucipir F 18 and were imaged for 20 min beginning 80 min after injection and again at 110 min after injection. Follow-up (retest) imaging occurred between 48 h and 4 wk after initial imaging. Images were spatially normalized to Montreal Neurological Institute template space. SUVRs were calculated using AAL (Automated Anatomical Labeling atlas) volumes of interest (VOIs) for parietal, temporal, occipital, anterior, and posterior hippocampal, parahippocampal, and fusiform regions, as well as a posterior neocortical VOI composed of average values from parietal, temporal, and occipital areas. Further, a VOI derived by discriminant analysis that maximally separated diagnostic groups (multiblock barycentric discriminant analysis [MUBADA]) was used. All VOIs were referenced to a subsection of cerebellar gray matter (cere-crus) as well as a parametrically derived white matter-based reference region (parametric estimate of reference signal intensity [PERSI]). t test, correlation analyses, and intraclass correlation coefficient were used to explore test-retest performance. Results: Test-retest analyses demonstrated low variability in flortaucipir F 18 SUVR. The SD of mean percentage change between test and retest using the PERSI reference region was 2.22% for a large posterior neocortical VOI, 1.84% for MUBADA, 1.46% for frontal, 1.98% for temporal, 2.28% for parietal, and 3.27% for occipital VOIs. Further, significant correlations (R2 > 0.85; P < 0.001) were observed for all regions, and intraclass correlation coefficient values (test-retest consistency) were greater than 0.92 for all regions. Conclusion: Significant test-retest reproducibility for flortaucipir F 18 was found across neocortical and mesial temporal lobe structures. These preliminary data suggest that flortaucipir F 18 tau imaging could be used to examine changes in tau burden over time.
Collapse
Affiliation(s)
- Michael D Devous
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| | - Abhinay D Joshi
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| | - Michael Navitsky
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| | - Sudeepti Southekal
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| | - Michael J Pontecorvo
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| | - Haiqing Shen
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| | - Ming Lu
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| | - William R Shankle
- Shankle Clinic Memory and Cognitive Disorders Program, Hoag Neurosciences Institute, Newport Beach, California; and
| | | | - Ken Marek
- Molecular NeuroImaging, New Haven, Connecticut
| | - Mark A Mintun
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia, Pennsylvania
| |
Collapse
|
36
|
Abstract
A compelling need in the field of neurodegenerative diseases is the development and validation of biomarkers for early identification and differential diagnosis. The availability of positron emission tomography (PET) neuroimaging tools for the assessment of molecular biology and neuropathology has opened new venues in the diagnostic design and the conduction of new clinical trials. PET techniques, allowing the in vivo assessment of brain function and pathology changes, are increasingly showing great potential in supporting clinical diagnosis also in the early and even preclinical phases of dementia. This review will summarize the most recent evidence on fluorine-18 fluorodeoxyglucose-, amyloid -, tau -, and neuroinflammation - PET tools, highlighting strengths and limitations and possible new perspectives in research and clinical applications. Appropriate use of PET tools is crucial for a prompt diagnosis and target evaluation of new developed drugs aimed at slowing or preventing dementia.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Arianna Sala
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Paola Caminiti
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
37
|
Van Dam D, Vermeiren Y, Dekker AD, Naudé PJW, Deyn PPD. Neuropsychiatric Disturbances in Alzheimer's Disease: What Have We Learned from Neuropathological Studies? Curr Alzheimer Res 2017; 13:1145-64. [PMID: 27137218 PMCID: PMC5070416 DOI: 10.2174/1567205013666160502123607] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/04/2016] [Accepted: 04/27/2016] [Indexed: 12/16/2022]
Abstract
Neuropsychiatric symptoms (NPS) are an integral part of the dementia syndrome and were therefore recently included in the core diagnostic criteria of dementia. The near universal prevalence of NPS in Alzheimer's disease (AD), combined with their disabling effects on patients and caregivers, is contrasted by the fact that few effective and safe treatments exist, which is in part to be attributed to our incomplete understanding of the neurobiology of NPS. In this review, we describe the pathological alterations typical for AD, including spreading and evolution of burden, effect on the molecular and cellular integrity, functional consequences and atrophy of NPS-relevant brain regions and circuits in correlation with specific NPS assessments. It is thereby clearly established that NPS are fundamental expressions of the underlying neurodegenerative brain disease and not simply reflect the patients' secondary response to their illness. Neuropathological studies, moreover, include a majority of end-stage patient samples, which may not correctly represent the pathophysiological environment responsible for particular NPS that may already be present in an early stage, or even prior to AD diagnosis. The burdensome nature and high prevalence of NPS, in combination with the absence of effective and safe pharmacotherapies, provide a strong incentive to continue neuropathological and neurochemical, as well as imaging and other relevant approaches to further improve our apprehension of the neurobiology of NPS.
Collapse
Affiliation(s)
| | | | | | | | - Peter P De Deyn
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, and, Faculty of Medical and Health Care Sciences, University of Antwerp, Universiteitsplein 1, BE-2610 Wilrijk (Antwerp), Belgium
| |
Collapse
|
38
|
Bischof GN, Endepols H, van Eimeren T, Drzezga A. Tau-imaging in neurodegeneration. Methods 2017; 130:114-123. [PMID: 28790016 DOI: 10.1016/j.ymeth.2017.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 10/19/2022] Open
Abstract
Pathological cerebral aggregations of proteins are suggested to play a crucial role in the development of neurodegenerative disorders. For example, aggregation of the protein ß-amyloid in form of extracellular amyloid-plaques as well as intraneuronal depositions of the protein tau in form of neurofibrillary tangles represent hallmarks of Alzheimer's disease (AD). Recently, novel tracers for in vivo molecular imaging of tau-aggregates in the brain have been introduced, complementing existing tracers for imaging amyloid-plaques. Available data on these novel tracers indicate that the subject of Tau-PET may be of considerable complexity. On the one hand this refers to the various forms of appearance of tau-pathology in different types of neurodegenerative disorders. On the other hand, a number of hurdles regarding validation of these tracers still need to be overcome with regard to comparability and standardization of the different tracers, observed off-target/non-specific binding and quantitative interpretation of the signal. These issues will have to be clarified before systematic clinical application of this exciting new methodological approach may become possible. Potential applications refer to early detection of neurodegeneration, differential diagnosis between tauopathies and non-tauopathies and specific patient selection and follow-up in therapy trials.
Collapse
Affiliation(s)
| | - Heike Endepols
- Department of Nuclear Medicine, University of Cologne, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany.
| |
Collapse
|
39
|
Coakeley S, Strafella AP. Imaging tau pathology in Parkinsonisms. NPJ Parkinsons Dis 2017; 3:22. [PMID: 28685158 PMCID: PMC5491530 DOI: 10.1038/s41531-017-0023-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 12/23/2022] Open
Abstract
The recent development of positron emission tomography radiotracers targeting pathological tau in vivo has led to numerous human trials. While investigations have primarily focused on the most common tauopathy, Alzheimer's disease, it is imperative that testing also be performed in parkinsonian tauopathies, such as progressive supranuclear palsy, corticobasal degeneration, and frontotemporal dementia and parkinsonism linked to chromosome 17. Tau aggregates differ in isoforms and conformations across disorders, and as a result one radiotracer may not be appropriate for all tauopathies. In this review, we evaluate the preclinical and clinical reports of current tau radiotracers in parkinsonian disorders. These radiotracers include [18F]FDDNP, [11C]PBB3, [18F]THK-5317, [18F]THK-5351, and [18F]AV-1451 ([18F]T807). There are concerns of off-target binding with [18F]FDDNP and [11C]PBB3, which may increase the signal to noise ratio and thereby decrease the efficacy of these radiotracers. Testing in [18F]THK-5317, [18F]THK-5351, and [18F]AV-1451 has been performed in progressive supranuclear palsy, while [18F]THK-5317 and [18F]AV-1451 have also been tested in corticobasal degeneration patients. [18F]THK-5317 and [18F]THK-5351 have demonstrated binding in brain regions known to be afflicted with pathological tau; however, due to small sample sizes these studies should be replicated before concluding their appropriateness in parkinsonian tauopathies. [18F]AV-1451 has demonstrated mixed results in progressive supranuclear palsy patients and post-mortem analysis shows minimal to no binding to non-Alzheimer's disease tauopathies brain slices.
Collapse
Affiliation(s)
- Sarah Coakeley
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON Canada
- Division of Brain, Imaging and Behaviour—Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Toronto, ON Canada
| | - Antonio P. Strafella
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON Canada
- Division of Brain, Imaging and Behaviour—Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Toronto, ON Canada
- Morton and Gloria Shulman Movement Disorder Unit and E.J. Safra Parkinson Disease Program, Neurology Division, Dept. of Medicine, Toronto Western Hospital, UHN, University of Toronto, Toronto, ON Canada
| |
Collapse
|
40
|
Gordon E, Rohrer JD, Fox NC. Advances in neuroimaging in frontotemporal dementia. J Neurochem 2017; 138 Suppl 1:193-210. [PMID: 27502125 DOI: 10.1111/jnc.13656] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/12/2022]
Abstract
Frontotemporal dementia (FTD) is a clinically and neuroanatomically heterogeneous neurodegenerative disorder with multiple underlying genetic and pathological causes. Whilst initial neuroimaging studies highlighted the presence of frontal and temporal lobe atrophy or hypometabolism as the unifying feature in patients with FTD, more detailed studies have revealed diverse patterns across individuals, with variable frontal or temporal predominance, differing degrees of asymmetry, and the involvement of other cortical areas including the insula and cingulate, as well as subcortical structures such as the basal ganglia and thalamus. Recent advances in novel imaging modalities including diffusion tensor imaging, resting-state functional magnetic resonance imaging and molecular positron emission tomography imaging allow the possibility of investigating alterations in structural and functional connectivity and the visualisation of pathological protein deposition. This review will cover the major imaging modalities currently used in research and clinical practice, focusing on the key insights they have provided into FTD, including the onset and evolution of pathological changes and also importantly their utility as biomarkers for disease detection and staging, differential diagnosis and measurement of disease progression. Validating neuroimaging biomarkers that are able to accomplish these tasks will be crucial for the ultimate goal of powering upcoming clinical trials by correctly stratifying patient enrolment and providing sensitive markers for evaluating the effects and efficacy of disease-modifying therapies. This review describes the key insights provided by research into the major neuroimaging modalities currently used in research and clinical practice, including what they tell us about the onset and evolution of FTD and how they may be used as biomarkers for disease detection and staging, differential diagnosis and measurement of disease progression. This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Elizabeth Gordon
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| |
Collapse
|
41
|
Canu E, Agosta F, Mandic-Stojmenovic G, Stojković T, Stefanova E, Inuggi A, Imperiale F, Copetti M, Kostic VS, Filippi M. Multiparametric MRI to distinguish early onset Alzheimer's disease and behavioural variant of frontotemporal dementia. NEUROIMAGE-CLINICAL 2017; 15:428-438. [PMID: 28616383 PMCID: PMC5458769 DOI: 10.1016/j.nicl.2017.05.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/12/2017] [Accepted: 05/25/2017] [Indexed: 12/11/2022]
Abstract
This prospective study explored whether an approach combining structural [cortical thickness and white matter (WM) microstructure] and resting state functional MRI can aid differentiation between 62 early onset Alzheimer's disease (EOAD) and 27 behavioural variant of frontotemporal dementia (bvFTD) patients. Random forest and receiver operator characteristic curve analyses assessed the ability of MRI in classifying the two clinical syndromes. All patients showed a distributed pattern of brain alterations relative to controls. Compared to bvFTD, EOAD patients showed bilateral inferior parietal cortical thinning and decreased default mode network functional connectivity. Compared to EOAD, bvFTD patients showed bilateral orbitofrontal and temporal cortical thinning, and WM damage of the corpus callosum, bilateral uncinate fasciculus, and left superior longitudinal fasciculus. Random forest analysis revealed that left inferior parietal cortical thickness (accuracy 0.78, specificity 0.76, sensitivity 0.83) and WM integrity of the right uncinate fasciculus (accuracy 0.81, specificity 0.96, sensitivity 0.43) were the best predictors of clinical diagnosis. The combination of cortical thickness and DT MRI measures was able to distinguish patients with EOAD and bvFTD with accuracy 0.82, specificity 0.76, and sensitivity 0.96. The diagnostic ability of MRI models was confirmed in a subsample of patients with biomarker-based clinical diagnosis. Multiparametric MRI is useful to identify brain alterations which are specific to EOAD and bvFTD. A severe cortical involvement is suggestive of EOAD, while a prominent WM damage is indicative of bvFTD. Multimodal MRI distinguishes in vivo EOAD and bvFTD patients EOAD and bvFTD show a distributed pattern of structural brain alterations A severe cortical involvement is suggestive of EOAD relative to bvFTD A prominent WM damage is indicative of bvFTD relative to EOAD
Collapse
Key Words
- ACE-R, Addenbrooke's Cognitive Examination-revised
- Behavioural variant of frontotemporal dementia
- CC, corpus callosum
- CSF, cerebrospinal fluid
- Cortical thickness
- DMN, default mode network
- DT, diffusion tensor
- Diagnosis
- EOAD, early onset Alzheimer's disease
- Early onset Alzheimer's disease
- GM, grey matter
- IC, independent component
- ILF, inferior longitudinal fasciculus
- LOAD, late onset Alzheimer's disease
- MNI, Montreal Neurological Institute
- NVI, Normalized Variable Importance
- RS fMRI, resting state functional MRI
- RSN, resting state network
- Resting state functional MRI
- SLF, superior longitudinal fasciculus
- TFCE, threshold-free cluster enhancement
- WM, white matter
- White matter (WM) damage
- bvFTD, behavioural variant frontotemporal dementia
Collapse
Affiliation(s)
- Elisa Canu
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Gorana Mandic-Stojmenovic
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Clinic of Neurology, Faculty of Medicine, University of Belgrade, Dr Subotića 6, PO Box 12, 11129 Belgrade 102, Serbia
| | - Tanja Stojković
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Dr Subotića 6, PO Box 12, 11129 Belgrade 102, Serbia
| | - Elka Stefanova
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Dr Subotića 6, PO Box 12, 11129 Belgrade 102, Serbia
| | - Alberto Inuggi
- Unit of Robotics, Brain and Cognitive Sciences, Istituto Italiano di Tecnologia, Via Morego, 30, 16163 Genoa, Italy
| | - Francesca Imperiale
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Massimiliano Copetti
- Biostatistics Unit, IRCCS-Ospedale Casa Sollievo della Sofferenza, Viale Cappuccini, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Vladimir S Kostic
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Dr Subotića 6, PO Box 12, 11129 Belgrade 102, Serbia
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy; Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
42
|
van Eimeren T, Bischof GN, Drzezga A. Is Tau Imaging More Than Just Upside-Down 18F-FDG Imaging? J Nucl Med 2017; 58:1357-1359. [DOI: 10.2967/jnumed.117.190082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/05/2017] [Indexed: 11/16/2022] Open
|
43
|
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the most common cause of dementia among the elderly population. The good correlation between the density and neocortical spread of neurofibrillary tangles (NFTs) and the severity of cognitive impairment offers an opportunity to use a noninvasive imaging technique such as positron emission tomography (PET) for early diagnosis and staging of the disease. PET imaging of NFTs holds promise not only as a diagnostic tool but also because it may enable the development of disease-modifying therapeutics for AD. In this review, we focus on the structural diversity of tau PET tracers, the challenges related to identifying high-affinity and highly selective NFT ligands, and recent progress in the clinical development of tau PET radioligands.
Collapse
Affiliation(s)
- Hartmuth C Kolb
- Janssen Research and Development, Neuroscience Biomarkers, San Diego, California 92121
| | - José Ignacio Andrés
- Janssen Research and Development, Discovery Sciences, Janssen-Cilag S.A., 45007 Toledo, Spain
| |
Collapse
|
44
|
Rombouts FJR, Andrés JI, Ariza M, Alonso JM, Austin N, Bottelbergs A, Chen L, Chupakhin V, Cleiren E, Fierens K, Fontana A, Langlois X, Leenaerts JE, Mariën J, Martínez Lamenca C, Salter R, Schmidt ME, Te Riele P, Wintmolders C, Trabanco AA, Zhang W, Macdonald G, Moechars D. Discovery of N-(Pyridin-4-yl)-1,5-naphthyridin-2-amines as Potential Tau Pathology PET Tracers for Alzheimer's Disease. J Med Chem 2017; 60:1272-1291. [PMID: 28106992 DOI: 10.1021/acs.jmedchem.6b01173] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A mini-HTS on 4000 compounds selected using 2D fragment-based similarity and 3D pharmacophoric and shape similarity to known selective tau aggregate binders identified N-(6-methylpyridin-2-yl)quinolin-2-amine 10 as a novel potent binder to human AD aggregated tau with modest selectivity versus aggregated β-amyloid (Aβ). Initial medicinal chemistry efforts identified key elements for potency and selectivity, as well as suitable positions for radiofluorination, leading to a first generation of fluoroalkyl-substituted quinoline tau binding ligands with suboptimal physicochemical properties. Further optimization toward a more optimal pharmacokinetic profile led to the discovery of 1,5-naphthyridine 75, a potent and selective tau aggregate binder with potential as a tau PET tracer.
Collapse
Affiliation(s)
- Frederik J R Rombouts
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - José-Ignacio Andrés
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Manuela Ariza
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - José Manuel Alonso
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Nigel Austin
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Astrid Bottelbergs
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Lu Chen
- Isotope Chemistry and Biotransformation, Janssen Research & Development , Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| | - Vladimir Chupakhin
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Erna Cleiren
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Katleen Fierens
- Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Alberto Fontana
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Xavier Langlois
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Joseph E Leenaerts
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Jonas Mariën
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Carolina Martínez Lamenca
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Rhys Salter
- Isotope Chemistry and Biotransformation, Janssen Research & Development , Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| | - Mark E Schmidt
- Neuroscience Experimental Medicine, Janssen Early Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Paula Te Riele
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Cindy Wintmolders
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Andrés A Trabanco
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag S. A. , C/Jarama 75A, 45007 Toledo, Spain
| | - Wei Zhang
- Neuroscience Biomarker Research, Janssen Research & Development , 3210 Merryfield Row, San Diego, California 92121, United States
| | - Gregor Macdonald
- Neuroscience Medicinal Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Dieder Moechars
- Neuroscience Biology, Janssen Research & Development, Janssen Pharmaceutica N. V. , Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
45
|
|
46
|
Stern RA, Tripodis Y, Baugh CM, Fritts NG, Martin BM, Chaisson C, Cantu RC, Joyce JA, Shah S, Ikezu T, Zhang J, Gercel-Taylor C, Taylor DD. Preliminary Study of Plasma Exosomal Tau as a Potential Biomarker for Chronic Traumatic Encephalopathy. J Alzheimers Dis 2016; 51:1099-109. [PMID: 26890775 PMCID: PMC4833534 DOI: 10.3233/jad-151028] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Chronic traumatic encephalopathy (CTE) is a tauopathy associated with prior exposure to repetitive head impacts, such as those incurred through American football and other collision sports. Diagnosis is made through neuropathological examination. Many of the clinical features of CTE are common in the general population, with and without a history of head impact exposure, making clinical diagnosis difficult. As is now common in the diagnosis of other neurodegenerative disorders, such as Alzheimer’s disease, there is a need for methods to diagnose CTE during life through objective biomarkers. Objective: The aim of this study was to examine tau-positive exosomes in plasma as a potential CTE biomarker. Methods: Subjects were 78 former National Football League (NFL) players and 16 controls. Extracellular vesicles were isolated from plasma. Fluorescent nanoparticle tracking analysis was used to determine the number of vesicles staining positive for tau. Results: The NFL group had higher exosomal tau than the control group (p < 0.0001). Exosomal tau discriminated between the groups, with 82% sensitivity, 100% specificity, 100% positive predictive value, and 53% negative predictive value. Within the NFL group, higher exosomal tau was associated with worse performance on tests of memory (p = 0.0126) and psychomotor speed (p = 0.0093). Conclusion: These preliminary findings suggest that exosomal tau in plasma may be an accurate, noninvasive CTE biomarker.
Collapse
Affiliation(s)
- Robert A Stern
- Boston University Alzheimer's Disease and CTE Center, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA.,Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease and CTE Center, Boston, MA, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Christine M Baugh
- Boston University Alzheimer's Disease and CTE Center, Boston, MA, USA
| | - Nathan G Fritts
- Boston University Alzheimer's Disease and CTE Center, Boston, MA, USA
| | - Brett M Martin
- Boston University Alzheimer's Disease and CTE Center, Boston, MA, USA.,Data Coordinating Center, Boston University School of Public Health, Boston, MA, USA
| | - Christine Chaisson
- Boston University Alzheimer's Disease and CTE Center, Boston, MA, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.,Data Coordinating Center, Boston University School of Public Health, Boston, MA, USA
| | - Robert C Cantu
- Boston University Alzheimer's Disease and CTE Center, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA.,Department of Neurosurgery, Emerson Hospital, Concord, MA, USA
| | | | | | - Tsuneya Ikezu
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jing Zhang
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
47
|
Ishiki A, Harada R, Okamura N, Tomita N, Rowe CC, Villemagne VL, Yanai K, Kudo Y, Arai H, Furumoto S, Tashiro M, Furukawa K. Tau imaging with [18F]THK-5351 in progressive supranuclear palsy. Eur J Neurol 2016; 24:130-136. [DOI: 10.1111/ene.13164] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/29/2016] [Indexed: 12/26/2022]
Affiliation(s)
- A. Ishiki
- Department of Geriatrics and Gerontology; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
| | - R. Harada
- Division of Neuro-imaging; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
| | - N. Okamura
- Department of Pharmacology; Tohoku University School of Medicine; Sendai Japan
- Division of Pharmacology; Faculty of Medicine; Tohoku Medical and Pharmaceutical University; Sendai Japan
| | - N. Tomita
- Department of Geriatrics and Gerontology; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
| | - C. C. Rowe
- Centre for PET; Austin Health; Heidelberg Victoria Australia
| | - V. L. Villemagne
- Centre for PET; Austin Health; Heidelberg Victoria Australia
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne Victoria Australia
| | - K. Yanai
- Department of Pharmacology; Tohoku University School of Medicine; Sendai Japan
| | - Y. Kudo
- Division of Neuro-imaging; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
| | - H. Arai
- Department of Geriatrics and Gerontology; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
| | - S. Furumoto
- Division of Radiopharmaceutical Chemistry; Cyclotron and Radioisotope Center; Tohoku University; Sendai Japan
| | - M. Tashiro
- Division of Cyclotron Nuclear Medicine; Cyclotron and Radioisotope Center; Tohoku University; Sendai Japan
| | - K. Furukawa
- Department of Geriatrics and Gerontology; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
- Division of Community of Medicine; Faculty of Medicine; Tohoku Medical and Pharmaceutical University; Sendai Japan
| |
Collapse
|
48
|
Okamura N, Harada R, Furukawa K, Furumoto S, Tago T, Yanai K, Arai H, Kudo Y. Advances in the development of tau PET radiotracers and their clinical applications. Ageing Res Rev 2016; 30:107-13. [PMID: 26802556 DOI: 10.1016/j.arr.2015.12.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 12/30/2015] [Accepted: 12/30/2015] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease and other neurodegenerative dementias belong to the family of tauopathies. These diseases are characterized by the deposition of insoluble tau aggregates possessing an enriched β-sheet structure. In vivo imaging of the tau deposits by positron emission tomography (PET) will facilitate the early and accurate diagnosis of these diseases, tracking of disease progression, assessment of disease severity, and prediction of disease prognosis. Furthermore, this technology is expected to play a vital role in the monitoring of treatment outcomes and in the selection of patients for the therapeutic trials of anti-dementia drugs. Recently, several tau PET tracers have been successfully developed and demonstrated as having high binding affinity and selectivity to tau protein deposits. Recent clinical studies using these tracers have demonstrated significant tracer retention in sites susceptible to tau deposition in Alzheimer's disease, as well as correlations with the disease severity and cognitive impairment in cases with dementia. These tracers, thus, have the potential to effectively diagnose the tauopathies. Further longitudinal assessment will clarify the effect of the tau deposition on the neurodegenerative process and cognitive decline and the interaction of tau with amyloid-β in the human brain.
Collapse
|
49
|
Turner RC, Lucke-Wold BP, Robson MJ, Lee JM, Bailes JE. Alzheimer's disease and chronic traumatic encephalopathy: Distinct but possibly overlapping disease entities. Brain Inj 2016; 30:1279-1292. [PMID: 27715315 PMCID: PMC5303562 DOI: 10.1080/02699052.2016.1193631] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 05/20/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE) have long been recognized as sharing some similar neuropathological features, mainly the presence of neurofibrilary tangles and hyperphosphorylated tau, but have generally been described as distinct entities. Evidence indicates that neurotrauma increases the risk of developing dementia and accelerates the progression of disease. Findings are emerging that CTE and AD may be present in the same patients. CLINICAL PRESENTATION This study presents a series of previously unpublished cases, with one case demonstrating possible neurotrauma-related AD, one pure CTE, and an example of a case exhibiting features of both AD and CTE. The future significance of this work lies not only in the confirmation of AD-CTE co-existence, but, more importantly, ways of generating a hypothesis about the possibility that CTE may accelerate AD development. Understanding the relationship between neurotrauma and neurodegenerative disease will help elucidate how distinct disease entities can co-exist in the same patient. It will ultimately require the use of pre-clinical animal models and repeat injury paradigms to investigate clinically relevant injury mechanisms. These models should produce a CTE-like phenotype that must be both neuropathologically and behaviourally similar to human disease. CONCLUSION This case series and review of the literature presents a discussion of AD and CTE in the context of neurotrauma. It highlights recent work from repetitive neurotrauma models with an emphasis on those exhibiting a CTE-like phenotype. Potential mechanisms of interest shared amongst AD and CTE are briefly addressed and future experiments are advocated for to enhance understanding of CTE pathophysiology and the relationship between CTE and AD.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Matthew J. Robson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John M. Lee
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| | - Julian E. Bailes
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| |
Collapse
|
50
|
Abstract
In vivo imaging of tau deposits is providing a better understanding of the temporal and spatial tau deposition in the brain, allowing a more comprehensive insight into the causes, diagnoses, and potentially treatment of tauopathies such as Alzheimer's disease, progressive supranuclear palsy, corticobasal syndrome, chronic traumatic encephalopathy, and some variants of frontotemporal lobar degeneration. The assessment of tau deposition in the brain over time will allow a deeper understanding of the relationship between tau and other variables such as cognition, genotype, and neurodegeneration, as well as assessing the role tau plays in ageing. Preliminary human studies suggest that tau imaging could also be used as a diagnostic, prognostic, and theranostic biomarker, as well as a surrogate marker for target engagement, patient recruitment, and efficacy monitoring for disease-specific therapeutic trials.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia; Department of Medicine, The University of Melbourne, Melbourne, Australia
| | | | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Melbourne, Australia; Department of Medicine, The University of Melbourne, Melbourne, Australia
| |
Collapse
|