1
|
Vacchi E, Ruiz-Barrio I, Melli G. Tau biomarkers for neurodegenerative diseases: Current state and perspectives. Parkinsonism Relat Disord 2025; 134:107772. [PMID: 40185651 DOI: 10.1016/j.parkreldis.2025.107772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative diseases, particularly tauopathies, pose significant global health challenges, especially in aging populations. Tauopathies are characterized by progressive neuronal damage and intracellular deposits of hyperphosphorylated tau. Early and accurate diagnosis is hindered by overlapping clinical features and reliance on post-mortem analyses, emphasizing the need for reliable in vivo biomarkers to improve early diagnosis and management. Advances in tau biomarkers and imaging have facilitated targeted Alzheimer's disease therapies, but progress for other tauopathies remains inadequate. Future diagnostic frameworks should integrate multiple biomarkers across different tissues within specific timelines. However, challenges such as co-pathologies and limited understanding of pathogenic mechanisms remain significant obstacles. Emerging ultrasensitive technologies, including seeding amplification assays and minimally invasive sources of biomarkers like skin biopsy, hold promise for biomarker discovery. Here, we present the current clinical classification of tau proteinopathies, the challenges that are posed by the actual diagnostic criteria, followed by the most recent advancements in tau biomarker technologies.
Collapse
Affiliation(s)
- Elena Vacchi
- Neurodegenerative Diseases Group, Laboratory for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Iñigo Ruiz-Barrio
- Department of Medicine, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain; Movement Disorders Unit, Neurology Department, Hospital de Sant Pau, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Giorgia Melli
- Neurodegenerative Diseases Group, Laboratory for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland; Neurology Department, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| |
Collapse
|
2
|
Magrath Guimet N, Falasco G, Bergamo Y, Urrutia L, Herrera JJ, Chrem Mendez P, Surace E, Allegri RF, Vazquez S, Bagnati PM. Behavioral variant frontotemporal dementia (bvFTD): PET biomarker characterization of metabolism (18F-FDG), amyloid (11C-PIB) and tau (18F-AV1451) and its clinical correlate - analysis of a cohort from Argentina. Alzheimers Dement 2025; 21:e70187. [PMID: 40271555 PMCID: PMC12019297 DOI: 10.1002/alz.70187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/07/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025]
Abstract
INTRODUCTION Imaging biomarkers are fundamental in diagnosing neurodegenerative diseases, but their use in FTD remains limited. This study examines PET biomarkers in Argentine bvFTD patients. METHODS We studied a cohort of bvFTD patients (n = 20) and controls (n = 21) with three different PET radiotracers (18F-FDG, 11C-PiB, and 18F-AV1451). RESULTS In bvFTD patients, 18F-FDG PET showed significant hypometabolism in frontotemporal regions, along with hypermetabolism in the precentral gyrus, compared to normal controls. 11C-PIB did not reveal a pattern typical of Alzheimer's disease, yet increased uptake was notably observed in the precentral region. We found 18F-AV1451 uptake in frontal lobe, parietal, precuneus, cuneus, posterior cingulum, highly significant in bvFTD with respect to NCs. DISCUSSION PET biomarkers are a crucial tool in diverse real-world clinical scenarios. However, their utility in revealing questions about the underlying pathology in FTD is still limited. HIGHLIGHTS First bvFTD study using 18F-FDG, 11C-PIB, and 18F-AV1451 PET in a Latin American cohort. Frontotemporal hypometabolism with compensatory precentral hypermetabolism due to amyloid. Amyloid deposits observed in the precentral gyrus without an Alzheimer's-like pattern. 18F-AV1451 shows limitations in specificity for bvFTD pathology. Study provides new insights into PET biomarker utility for bvFTD clinical assessment.
Collapse
Affiliation(s)
- Nahuel Magrath Guimet
- Department of Cognitive Neurology, Neuropsychiatry and NeuropsychologyFleniBuenos AiresArgentina
- Atlantic Fellow for Equity in Brain Health, Global Brain Health Institute (GBHI), University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Germán Falasco
- Molecular Imaging CenterFleniProvincia de Buenos AiresArgentina
| | - Yanina Bergamo
- Molecular Imaging CenterFleniProvincia de Buenos AiresArgentina
| | - Leandro Urrutia
- Molecular Imaging CenterFleniProvincia de Buenos AiresArgentina
| | - Julio Jose Herrera
- Department of Cognitive Neurology, Neuropsychiatry and NeuropsychologyFleniBuenos AiresArgentina
| | - Patricio Chrem Mendez
- Department of Cognitive Neurology, Neuropsychiatry and NeuropsychologyFleniBuenos AiresArgentina
| | - Ezequiel Surace
- Laboratory of Neurodegenerative DiseasesInstitute of Neuroscience‐Fleni (CONICET)Buenos AiresArgentina
| | - Ricardo Francisco Allegri
- Department of Cognitive Neurology, Neuropsychiatry and NeuropsychologyFleniBuenos AiresArgentina
- Neurosciences DepartmentUniversidad de La Costa. Cl, AtlánticoBarranquillaColombia
| | - Silvia Vazquez
- Molecular Imaging CenterFleniProvincia de Buenos AiresArgentina
| | - Pablo Miguel Bagnati
- Department of Cognitive Neurology, Neuropsychiatry and NeuropsychologyFleniBuenos AiresArgentina
| |
Collapse
|
3
|
Horie K, Salvadó G, Koppisetti RK, Janelidze S, Barthélemy NR, He Y, Sato C, Gordon BA, Jiang H, Benzinger TLS, Stomrud E, Holtzman DM, Mattsson-Carlgren N, Morris JC, Palmqvist S, Ossenkoppele R, Schindler SE, Hansson O, Bateman RJ. Plasma MTBR-tau243 biomarker identifies tau tangle pathology in Alzheimer's disease. Nat Med 2025:10.1038/s41591-025-03617-7. [PMID: 40164726 DOI: 10.1038/s41591-025-03617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
Insoluble tau aggregates within neurofibrillary tangles are a defining neuropathological feature of Alzheimer's disease (AD) and closely correlate with clinical symptoms. Although tau pathology can be assessed using tau positron emission tomography, a more accessible biomarker is needed for diagnosis, prognosis and tracking treatment effects. Here we present a new plasma tau species, the endogenously cleaved, microtubule-binding region containing residue 243 (eMTBR-tau243), which specifically reflects tau tangle pathology. Across the AD spectrum in three different cohorts (n = 108, 55 and 739), plasma eMTBR-tau243 levels were significantly elevated at the mild cognitive impairment stage and increased further in dementia. Plasma eMTBR-tau243 showed strong associations with tau positron emission tomography binding (β = 0.72, R2 = 0.56) and cognitive performance (β = 0.60, R2 = 0.40), outperforming other plasma tau (%p-tau217 and %p-tau205) biomarkers. These results suggest that plasma eMTBR-tau243 may be useful for estimating the tauopathy load in AD, thereby improving the diagnostic evaluation of AD in clinical practice and monitoring the efficacy of tau-targeted therapies in clinical trials.
Collapse
Affiliation(s)
- Kanta Horie
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Eisai Inc., Nutley, NJ, USA.
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rama K Koppisetti
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Nicolas R Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chihiro Sato
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| | - Randall J Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Qi C, Lövestam S, Murzin AG, Peak-Chew S, Franco C, Bogdani M, Latimer C, Murrell JR, Cullinane PW, Jaunmuktane Z, Bird TD, Ghetti B, Scheres SHW, Goedert M. Tau filaments with the Alzheimer fold in human MAPT mutants V337M and R406W. Nat Struct Mol Biol 2025:10.1038/s41594-025-01498-5. [PMID: 40044789 DOI: 10.1038/s41594-025-01498-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/23/2025] [Indexed: 03/12/2025]
Abstract
Frontotemporal dementia (FTD) and Alzheimer's disease (AD) are the most common forms of early-onset dementia. Unlike AD, FTD begins with behavioral changes before the development of cognitive impairment. Dominantly inherited mutations in MAPT, the microtubule-associated protein tau gene, give rise to cases of FTD and parkinsonism linked to chromosome 17. These individuals develop abundant filamentous tau inclusions in brain cells in the absence of β-amyloid deposits. Here, we used cryo-electron microscopy to determine the structures of tau filaments from the brains of human MAPT mutants V337M and R406W. Both amino acid substitutions gave rise to tau filaments with the Alzheimer fold, which consisted of paired helical filaments in all V337M and R406W cases and of straight filaments in two V337M cases. We also identified another assembly of the Alzheimer fold into triple tau filaments in a V337M case. Filaments assembled from recombinant tau (297-391) with substitution V337M had the Alzheimer fold and showed an increased rate of assembly.
Collapse
Affiliation(s)
- Chao Qi
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | | | | | | | - Marika Bogdani
- Departments of Neurology and Pathology, University of Washington, Seattle, WA, USA
- Veterans Administration Puget Sound Health Care System, Seattle, WA, USA
| | - Caitlin Latimer
- Departments of Neurology and Pathology, University of Washington, Seattle, WA, USA
- Veterans Administration Puget Sound Health Care System, Seattle, WA, USA
| | - Jill R Murrell
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick W Cullinane
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College, London, UK
- Queen Square Brain Bank for Neurological Disorders, Institute of Neurology, University College, London, UK
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College, London, UK
- Queen Square Brain Bank for Neurological Disorders, Institute of Neurology, University College, London, UK
| | - Thomas D Bird
- Departments of Neurology and Pathology, University of Washington, Seattle, WA, USA
- Veterans Administration Puget Sound Health Care System, Seattle, WA, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | |
Collapse
|
5
|
Wang L, Sooram B, Kumar R, Schedin-Weiss S, Tjernberg LO, Winblad B. Tau degradation in Alzheimer's disease: Mechanisms and therapeutic opportunities. Alzheimers Dement 2025; 21:e70048. [PMID: 40109019 PMCID: PMC11923393 DOI: 10.1002/alz.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/22/2025]
Abstract
In Alzheimer's disease (AD), tau undergoes abnormal post-translational modifications and aggregations. Impaired intracellular degradation pathways further exacerbate the accumulation of pathological tau. A new strategy - targeted protein degradation - recently emerged as a modality in drug discovery where bifunctional molecules bring the target protein close to the degradation machinery to promote clearance. Since 2016, this strategy has been applied to tau pathologies and attracted broad interest in academia and the pharmaceutical industry. However, a systematic review of recent studies on tau degradation mechanisms is lacking. Here we review tau degradation mechanisms (the ubiquitin-proteasome system and the autophagy-lysosome pathway), their dysfunction in AD, and tau-targeted degraders, such as proteolysis-targeting chimeras and autophagy-targeting chimeras. We emphasize the need for a continuous exploration of tau degradation mechanisms and provide a future perspective for developing tau-targeted degraders, encouraging researchers to work on new treatment options for AD patients. HIGHLIGHTS: Post-translational modifications, aggregation, and mutations affect tau degradation. A vicious circle exists between impaired degradation pathways and tau pathologies. Ubiquitin plays an important role in complex degradation pathways. Tau-targeted degraders provide promising strategies for novel AD treatment.
Collapse
Affiliation(s)
- Lisha Wang
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Banesh Sooram
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Rajnish Kumar
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, India
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
6
|
Schweighauser M, Shi Y, Murzin AG, Garringer HJ, Vidal R, Murrell JR, Erro ME, Seelaar H, Ferrer I, van Swieten JC, Ghetti B, Scheres SH, Goedert M. Novel tau filament folds in individuals with MAPT mutations P301L and P301T. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608062. [PMID: 39185206 PMCID: PMC11343192 DOI: 10.1101/2024.08.15.608062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Mutations in MAPT, the microtubule-associated protein tau gene, give rise to cases of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) with abundant filamentous tau inclusions in brain cells. Individuals with pathological MAPT variants exhibit behavioural changes, cognitive impairment and signs of parkinsonism. Missense mutations of residue P301, which are the most common MAPT mutations associated with FTDP-17, give rise to the assembly of mutant four-repeat tau into filamentous inclusions, in the absence of extracellular deposits. Here we report the cryo-EM structures of tau filaments from five individuals belonging to three unrelated families with mutation P301L and from one individual belonging to a family with mutation P301T. A novel three-lobed tau fold resembling the two-layered tau fold of Pick's disease was present in all cases with the P301L tau mutation. Two different tau folds were found in the case with mutation P301T, the less abundant of which was a variant of the three-lobed fold. The major P301T tau fold was V-shaped, with partial similarity to the four-layered tau folds of corticobasal degeneration and argyrophilic grain disease. These findings suggest that FTDP-17 with mutations in P301 should be considered distinct inherited tauopathies and that model systems with these mutations should be used with caution in the study of sporadic tauopathies.
Collapse
Affiliation(s)
- Manuel Schweighauser
- MRC Laboratory of Molecular Biology, Cambridge, UK
- These authors contributed equally
| | - Yang Shi
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Pathology of the First Affiliated Hospital and School of Brain Science, Zhejiang University, Hangzhou, China
- These authors contributed equally
| | - Alexey G. Murzin
- MRC Laboratory of Molecular Biology, Cambridge, UK
- These authors contributed equally
| | - Holly J. Garringer
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Jill R. Murrell
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of the University of Pennsylvania, Philadelphia, USA
| | - M. Elena Erro
- Department of Neurology, Hospital Universitario de Navarra, Brain Bank NavarraBiomed, Pamplona, Spain
| | - Harro Seelaar
- Department of Neurology, Erasmus University, Rotterdam, The Netherlands
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Spain
| | | | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Sjors H.W. Scheres
- MRC Laboratory of Molecular Biology, Cambridge, UK
- These authors jointly supervised the work
| | - Michel Goedert
- MRC Laboratory of Molecular Biology, Cambridge, UK
- These authors jointly supervised the work
| |
Collapse
|
7
|
Lee J, Burkett BJ, Min HK, Senjem ML, Dicks E, Corriveau-Lecavalier N, Mester CT, Wiste HJ, Lundt ES, Murray ME, Nguyen AT, Reichard RR, Botha H, Graff-Radford J, Barnard LR, Gunter JL, Schwarz CG, Kantarci K, Knopman DS, Boeve BF, Lowe VJ, Petersen RC, Jack CR, Jones DT. Synthesizing images of tau pathology from cross-modal neuroimaging using deep learning. Brain 2024; 147:980-995. [PMID: 37804318 PMCID: PMC10907092 DOI: 10.1093/brain/awad346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/30/2023] [Accepted: 09/24/2023] [Indexed: 10/09/2023] Open
Abstract
Given the prevalence of dementia and the development of pathology-specific disease-modifying therapies, high-value biomarker strategies to inform medical decision-making are critical. In vivo tau-PET is an ideal target as a biomarker for Alzheimer's disease diagnosis and treatment outcome measure. However, tau-PET is not currently widely accessible to patients compared to other neuroimaging methods. In this study, we present a convolutional neural network (CNN) model that imputes tau-PET images from more widely available cross-modality imaging inputs. Participants (n = 1192) with brain T1-weighted MRI (T1w), fluorodeoxyglucose (FDG)-PET, amyloid-PET and tau-PET were included. We found that a CNN model can impute tau-PET images with high accuracy, the highest being for the FDG-based model followed by amyloid-PET and T1w. In testing implications of artificial intelligence-imputed tau-PET, only the FDG-based model showed a significant improvement of performance in classifying tau positivity and diagnostic groups compared to the original input data, suggesting that application of the model could enhance the utility of the metabolic images. The interpretability experiment revealed that the FDG- and T1w-based models utilized the non-local input from physically remote regions of interest to estimate the tau-PET, but this was not the case for the Pittsburgh compound B-based model. This implies that the model can learn the distinct biological relationship between FDG-PET, T1w and tau-PET from the relationship between amyloid-PET and tau-PET. Our study suggests that extending neuroimaging's use with artificial intelligence to predict protein specific pathologies has great potential to inform emerging care models.
Collapse
Affiliation(s)
- Jeyeon Lee
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biomedical Engineering, Hanyang University, Seoul 04763, Korea
| | - Brian J Burkett
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hoon-Ki Min
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ellen Dicks
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Carly T Mester
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Heather J Wiste
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Emily S Lundt
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ross R Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David T Jones
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Badihian N, Ali F, Botha H, Savica R, Machulda MM, Clark HM, Stierwalt JAG, Pham NTT, Baker MC, Rademakers R, Lowe V, Whitwell JL, Josephs KA. The MAPT p.E342K and p.R406W mutations are associated with progressive supranuclear palsy with atypical features. Parkinsonism Relat Disord 2024; 119:105962. [PMID: 38134678 PMCID: PMC10842922 DOI: 10.1016/j.parkreldis.2023.105962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION Progressive supranuclear palsy (PSP) is an atypical parkinsonism caused by the intracerebral aggregation of the microtubule-associated protein tau (MAPT) which is encoded by MAPT gene. Although PSP is a sporadic disease, MAPT mutations have been reported in rare cases. METHODS Among 190 patients with PSP who were recruited by the Neurodegenerative Research Group at Mayo Clinic during 2009-2023, we identified two patients who fulfilled diagnostic criteria for PSP-Richardson's syndrome (PSP-RS) and harbor novel MAPT mutations. To better investigate the potential effects of these mutations, we compared the clinical, and neuroimaging characteristics of these two patients to 20 randomly selected patients with PSP-RS without a MAPT mutation. RESULTS MAPT c.1024G > A, p. Glu342Lys, and MAPT c.1217 G > A, p. Arg406Gln mutations were found in 2 men who developed PSP-RS with atypical features at the ages of 60 and 62 years, respectively. Glu342Lys mutation was associated with features resembling alpha-synucleinopathies (autonomic dysfunction, dream enactment behavior), while both mutations were associated with features suggestive of Alzheimer's disease with poorer performance on tests of episodic memory. Comparison of 18F-flortaucipir uptake between the two MAPT mutation cases with 20 patients without a mutation revealed increased signal on flortaucipir-PET in bilateral medial temporal lobe regions (amygdala, entorhinal cortices, hippocampus, parahippocampus) but not in PSP-related regions (globus pallidum, midbrain, superior frontal cortex and dentate nucleus of the cerebellum). CONCLUSION Glu342Lys and Arg406Gln mutations appear to modify the PSP-RS phenotype by targeting the medial temporal lobe regions resulting in more memory loss and greater flortaucipir uptake.
Collapse
Affiliation(s)
- Negin Badihian
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Farwa Ali
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Matthew C Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Rosa Rademakers
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
9
|
Corriveau-Lecavalier N, Barnard LR, Przybelski SA, Gogineni V, Botha H, Graff-Radford J, Ramanan VK, Forsberg LK, Fields JA, Machulda MM, Rademakers R, Gavrilova RH, Lapid MI, Boeve BF, Knopman DS, Lowe VJ, Petersen RC, Jack CR, Kantarci K, Jones DT. Assessing network degeneration and phenotypic heterogeneity in genetic frontotemporal lobar degeneration by decoding FDG-PET. Neuroimage Clin 2023; 41:103559. [PMID: 38147792 PMCID: PMC10944211 DOI: 10.1016/j.nicl.2023.103559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023]
Abstract
Genetic mutations causative of frontotemporal lobar degeneration (FTLD) are highly predictive of a specific proteinopathy, but there exists substantial inter-individual variability in their patterns of network degeneration and clinical manifestations. We collected clinical and 18Fluorodeoxyglucose-positron emission tomography (FDG-PET) data from 39 patients with genetic FTLD, including 11 carrying the C9orf72 hexanucleotide expansion, 16 carrying a MAPT mutation and 12 carrying a GRN mutation. We performed a spectral covariance decomposition analysis between FDG-PET images to yield unbiased latent patterns reflective of whole brain patterns of metabolism ("eigenbrains" or EBs). We then conducted linear discriminant analyses (LDAs) to perform EB-based predictions of genetic mutation and predominant clinical phenotype (i.e., behavior/personality, language, asymptomatic). Five EBs were significant and explained 58.52 % of the covariance between FDG-PET images. EBs indicative of hypometabolism in left frontotemporal and temporo-parietal areas distinguished GRN mutation carriers from other genetic mutations and were associated with predominant language phenotypes. EBs indicative of hypometabolism in prefrontal and temporopolar areas with a right hemispheric predominance were mostly associated with predominant behavioral phenotypes and distinguished MAPT mutation carriers from other genetic mutations. The LDAs yielded accuracies of 79.5 % and 76.9 % in predicting genetic status and predominant clinical phenotype, respectively. A small number of EBs explained a high proportion of covariance in patterns of network degeneration across FTLD-related genetic mutations. These EBs contained biological information relevant to the variability in the pathophysiological and clinical aspects of genetic FTLD, and for offering valuable guidance in complex clinical decision-making, such as decisions related to genetic testing.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic Rochester, USA; Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | | | | | | | - Hugo Botha
- Department of Neurology, Mayo Clinic Rochester, USA
| | | | | | | | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic Jacksonville, USA; VIB-UA Center for Molecular Neurology, VIB, University of Antwerp, Belgium
| | | | - Maria I Lapid
- Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic Rochester, USA
| | | | | | | | - David T Jones
- Department of Neurology, Mayo Clinic Rochester, USA; Department of Radiology, Mayo Clinic Rochester, USA.
| |
Collapse
|
10
|
Burnham SC, Iaccarino L, Pontecorvo MJ, Fleisher AS, Lu M, Collins EC, Devous MD. A review of the flortaucipir literature for positron emission tomography imaging of tau neurofibrillary tangles. Brain Commun 2023; 6:fcad305. [PMID: 38187878 PMCID: PMC10768888 DOI: 10.1093/braincomms/fcad305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease is defined by the presence of β-amyloid plaques and neurofibrillary tau tangles potentially preceding clinical symptoms by many years. Previously only detectable post-mortem, these pathological hallmarks are now identifiable using biomarkers, permitting an in vivo definitive diagnosis of Alzheimer's disease. 18F-flortaucipir (previously known as 18F-T807; 18F-AV-1451) was the first tau positron emission tomography tracer to be introduced and is the only Food and Drug Administration-approved tau positron emission tomography tracer (Tauvid™). It has been widely adopted and validated in a number of independent research and clinical settings. In this review, we present an overview of the published literature on flortaucipir for positron emission tomography imaging of neurofibrillary tau tangles. We considered all accessible peer-reviewed literature pertaining to flortaucipir through 30 April 2022. We found 474 relevant peer-reviewed publications, which were organized into the following categories based on their primary focus: typical Alzheimer's disease, mild cognitive impairment and pre-symptomatic populations; atypical Alzheimer's disease; non-Alzheimer's disease neurodegenerative conditions; head-to-head comparisons with other Tau positron emission tomography tracers; and technical considerations. The available flortaucipir literature provides substantial evidence for the use of this positron emission tomography tracer in assessing neurofibrillary tau tangles in Alzheimer's disease and limited support for its use in other neurodegenerative disorders. Visual interpretation and quantitation approaches, although heterogeneous, mostly converge and demonstrate the high diagnostic and prognostic value of flortaucipir in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Ming Lu
- Avid, Eli Lilly and Company, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
11
|
Horie K, Salvadó G, Barthélemy NR, Janelidze S, Li Y, He Y, Saef B, Chen CD, Jiang H, Strandberg O, Pichet Binette A, Palmqvist S, Sato C, Sachdev P, Koyama A, Gordon BA, Benzinger TLS, Holtzman DM, Morris JC, Mattsson-Carlgren N, Stomrud E, Ossenkoppele R, Schindler SE, Hansson O, Bateman RJ. CSF MTBR-tau243 is a specific biomarker of tau tangle pathology in Alzheimer's disease. Nat Med 2023; 29:1954-1963. [PMID: 37443334 PMCID: PMC10427417 DOI: 10.1038/s41591-023-02443-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023]
Abstract
Aggregated insoluble tau is one of two defining features of Alzheimer's disease. Because clinical symptoms are strongly correlated with tau aggregates, drug development and clinical diagnosis need cost-effective and accessible specific fluid biomarkers of tau aggregates; however, recent studies suggest that the fluid biomarkers currently available cannot specifically track tau aggregates. We show that the microtubule-binding region (MTBR) of tau containing the residue 243 (MTBR-tau243) is a new cerebrospinal fluid (CSF) biomarker specific for insoluble tau aggregates and compared it to multiple other phosphorylated tau measures (p-tau181, p-tau205, p-tau217 and p-tau231) in two independent cohorts (BioFINDER-2, n = 448; and Knight Alzheimer Disease Research Center, n = 219). MTBR-tau243 was most strongly associated with tau-positron emission tomography (PET) and cognition, whereas showing the lowest association with amyloid-PET. In combination with p-tau205, MTBR-tau243 explained most of the total variance in tau-PET burden (0.58 ≤ R2 ≤ 0.75) and the performance in predicting cognitive measures (0.34 ≤ R2 ≤ 0.48) approached that of tau-PET (0.44 ≤ R2 ≤ 0.52). MTBR-tau243 levels longitudinally increased with insoluble tau aggregates, unlike CSF p-tau species. CSF MTBR-tau243 is a specific biomarker of tau aggregate pathology, which may be utilized in interventional trials and in the diagnosis of patients. Based on these findings, we propose to revise the A/T/(N) criteria to include MTBR-tau243 as representing insoluble tau aggregates ('T').
Collapse
Grants
- P30 AG066444 NIA NIH HHS
- R01 AG070941 NIA NIH HHS
- P01 AG003991 NIA NIH HHS
- P01 AG026276 NIA NIH HHS
- P30 NS048056 NINDS NIH HHS
- S10 OD025214 NIH HHS
- The Tracy Family SILQ Center established by the Tracy Family, Richard Frimel and Gary Werths, GHR Foundation, David Payne, and the Willman Family brought together by The Foundation for Barnes-Jewish Hospital.
- Eisai industry grant
- The European Union’s Horizon 2020 research and innovation program under the Marie Sklodowska-Curie action grant agreement No 101061836, from Greta och Johan Kocks research grants and, travel grants from the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- The Swedish Research Council (2016-00906), the Knut and Alice Wallenberg foundation (2017-0383), the Marianne and Marcus Wallenberg foundation (2015.0125), the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University, the Swedish Alzheimer Foundation (AF-939932), the Swedish Brain Foundation (FO2021-0293), The Parkinson foundation of Sweden (1280/20), the Cure Alzheimer’s fund, the Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, the Skåne University Hospital Foundation (2020-O000028), Regionalt Forskningsstöd (2020-0314) and the Swedish federal government under the ALF agreement (2018-Projekt0279)
- The Knight ADRC developmental project
Collapse
Affiliation(s)
- Kanta Horie
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Eisai Inc., Nutley, NJ, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Nicolas R Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Benjamin Saef
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Chihiro Sato
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| | - Randall J Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
12
|
Antonioni A, Raho EM, Lopriore P, Pace AP, Latino RR, Assogna M, Mancuso M, Gragnaniello D, Granieri E, Pugliatti M, Di Lorenzo F, Koch G. Frontotemporal Dementia, Where Do We Stand? A Narrative Review. Int J Mol Sci 2023; 24:11732. [PMID: 37511491 PMCID: PMC10380352 DOI: 10.3390/ijms241411732] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease of growing interest, since it accounts for up to 10% of middle-age-onset dementias and entails a social, economic, and emotional burden for the patients and caregivers. It is characterised by a (at least initially) selective degeneration of the frontal and/or temporal lobe, generally leading to behavioural alterations, speech disorders, and psychiatric symptoms. Despite the recent advances, given its extreme heterogeneity, an overview that can bring together all the data currently available is still lacking. Here, we aim to provide a state of the art on the pathogenesis of this disease, starting with established findings and integrating them with more recent ones. In particular, advances in the genetics field will be examined, assessing them in relation to both the clinical manifestations and histopathological findings, as well as considering the link with other diseases, such as amyotrophic lateral sclerosis (ALS). Furthermore, the current diagnostic criteria will be explored, including neuroimaging methods, nuclear medicine investigations, and biomarkers on biological fluids. Of note, the promising information provided by neurophysiological investigations, i.e., electroencephalography and non-invasive brain stimulation techniques, concerning the alterations in brain networks and neurotransmitter systems will be reviewed. Finally, current and experimental therapies will be considered.
Collapse
Affiliation(s)
- Annibale Antonioni
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
- Doctoral Program in Translational Neurosciences and Neurotechnologies, University of Ferrara, 44121 Ferrara, Italy
| | - Emanuela Maria Raho
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Piervito Lopriore
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonia Pia Pace
- Institute of Radiology, Department of Medicine, University of Udine, University Hospital S. Maria della Misericordia, Azienda Sanitaria-Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Raffaela Rita Latino
- Complex Structure of Neurology, Emergency Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Martina Assogna
- Centro Demenze, Policlinico Tor Vergata, University of Rome 'Tor Vergata', 00133 Rome, Italy
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Michelangelo Mancuso
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Daniela Gragnaniello
- Nuerology Unit, Neurosciences and Rehabilitation Department, Ferrara University Hospital, 44124 Ferrara, Italy
| | - Enrico Granieri
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Maura Pugliatti
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Francesco Di Lorenzo
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
- Iit@Unife Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Section of Human Physiology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
13
|
Conte M, De Feo MS, Sidrak MMA, Corica F, Gorica J, Granese GM, Filippi L, De Vincentis G, Frantellizzi V. Imaging of Tauopathies with PET Ligands: State of the Art and Future Outlook. Diagnostics (Basel) 2023; 13:diagnostics13101682. [PMID: 37238166 DOI: 10.3390/diagnostics13101682] [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: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: Tauopathies are a group of diseases characterized by the deposition of abnormal tau protein. They are distinguished into 3R, 4R, and 3R/4R tauopathies and also include Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Positron emission tomography (PET) imaging represents a pivotal instrument to guide clinicians. This systematic review aims to summarize the current and novel PET tracers. (2) Methods: Literature research was conducted on Pubmed, Scopus, Medline, Central, and the Web of Science using the query "pet ligands" and "tauopathies". Articles published from January 2018 to 9 February, 2023, were searched. Only studies on the development of novel PET radiotracers for imaging in tauopathies or comparative studies between existing PET tracers were included. (3) Results: A total of 126 articles were found, as follows: 96 were identified from PubMed, 27 from Scopus, one on Central, two on Medline, and zero on the Web of Science. Twenty-four duplicated works were excluded, and 63 articles did not satisfy the inclusion criteria. The remaining 40 articles were included for quality assessment. (4) Conclusions: PET imaging represents a valid instrument capable of helping clinicians in diagnosis, but it is not always perfect in differential diagnosis, even if further investigations on humans for novel promising ligands are needed.
Collapse
Affiliation(s)
- Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Silvia De Feo
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marko Magdi Abdou Sidrak
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Ferdinando Corica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Joana Gorica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Giorgia Maria Granese
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Luca Filippi
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, 00410 Latina, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
14
|
Abstract
Brain PET adds value in diagnosing neurodegenerative disorders, especially frontotemporal dementia (FTD) due to its syndromic presentation that overlaps with a variety of other neurodegenerative and psychiatric disorders. 18F-FDG-PET has improved sensitivity and specificity compared with structural MR imaging, with optimal diagnostic results achieved when both techniques are utilized. PET demonstrates superior sensitivity compared with SPECT for FTD diagnosis that is primarily a supplement to other imaging and clinical evaluations. Tau-PET and amyloid-PET primary use in FTD diagnosis is differentiation from Alzheimer disease, although these methods are limited mainly to research settings.
Collapse
Affiliation(s)
- Joshua Ward
- Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in Saint. Louis, Saint Louis, MO 63130, USA
| | - Maria Ly
- Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in Saint. Louis, Saint Louis, MO 63130, USA
| | - Cyrus A. Raji
- Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in Saint. Louis, Saint Louis, MO 63130, USA,Department of Neurology, Washington University in St. Louis, 4525 Scott Avenue, St. Louis, MO 63110, USA,Corresponding author. Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in Saint. Louis, Saint Louis, MO 63130.
| |
Collapse
|
15
|
Tang Y, Li L, Hu T, Jiao F, Han L, Li S, Xu Z, Fan Y, Sun Y, Liu F, Yen TC, Zuo C, Wang J. In Vivo 18 F-Florzolotau Tau Positron Emission Tomography Imaging in Parkinson's Disease Dementia. Mov Disord 2023; 38:147-152. [PMID: 36368769 DOI: 10.1002/mds.29273] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/19/2022] [Accepted: 10/30/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Tau pathology is observed during autopsy in many patients with Parkinson's disease dementia (PDD). Positron emission tomography (PET) imaging using the tracer 18 F-florzolotau has the potential to capture tau accumulation in the living brain. OBJECTIVE The aim was to describe the results of 18 F-florzolotau PET/CT (computed tomography) imaging in patients with PDD. METHODS Ten patients with PDD, 9 with Parkinson's disease with normal cognition (PD-NC), and 9 age-matched healthy controls (HCs) were enrolled. Clinical assessments and 18 F-florzolotau PET/CT imaging were performed. RESULTS 18 F-Florzolotau uptake was significantly higher in the cortical regions of patients with PDD compared with both PD-NC and HCs, especially in the temporal lobe. Notably, 18 F-florzolotau uptake in the occipital lobe of patients with PDD showed a significant correlation with cognitive impairment as reflected by Mini-Mental State Examination (MMSE) scores. CONCLUSIONS 18 F-Florzolotau PET imaging can effectively capture the occurrence of tau pathology in patients with PDD, which was also linked to MMSE scores. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yilin Tang
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ling Li
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Tianyu Hu
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangyang Jiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Linlin Han
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shiyu Li
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiheng Xu
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yun Fan
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yimin Sun
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fengtao Liu
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Neurology and National Research Center for Aging and Medicine and National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Josephs KA, Tosakulwong N, Gatto RG, Weigand SD, Ali F, Botha H, Graff‐Radford J, Machulda MM, Savica R, Schwarz CG, Senjem ML, Boeve BF, Kantarci K, Jones DT, Ramanan VK, Fields JA, Reichard RR, Dickson DW, Petersen RC, Jack CR, Lowe VJ, Whitwell JL. Optimum Differentiation of Frontotemporal Lobar Degeneration from Alzheimer Disease Achieved with Cross-Sectional Tau Positron Emission Tomography. Ann Neurol 2022; 92:1016-1029. [PMID: 36054427 PMCID: PMC9804568 DOI: 10.1002/ana.26479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVE This study was undertaken to assess cross-sectional and longitudinal [18 F]-flortaucipir positron emission tomography (PET) uptake in pathologically confirmed frontotemporal lobar degeneration (FTLD) and to compare FTLD to cases with high and low levels of Alzheimer disease (AD) neuropathologic changes (ADNC). METHODS One hundred forty-three participants who had completed at least one flortaucipir PET and had autopsy-confirmed FTLD (n = 52) or high (n = 58) or low ADNC (n = 33) based on Braak neurofibrillary tangle stages 0-IV versus V-VI were included. Flortaucipir standard uptake value ratios (SUVRs) were calculated for 9 regions of interest (ROIs): an FTLD meta-ROI, midbrain, globus pallidum, an AD meta-ROI, entorhinal, inferior temporal, orbitofrontal, precentral, and medial parietal. Linear mixed effects models were used to compare mean baseline SUVRs and annual rate of change in SUVR by group. Sensitivity and specificity to distinguish FTLD from high and low ADNC were calculated. RESULTS Baseline uptake in the FTLD meta-ROI, midbrain, and globus pallidus was greater in FTLD than high and low ADNC. No region showed a greater rate of flortaucipir accumulation in FTLD. Baseline uptake in the AD-related regions and orbitofrontal and precentral cortices was greater in high ADNC, and all showed greater rates of accumulation compared to FTLD. Baseline differences were superior to longitudinal rates in differentiating FTLD from high and low ADNC. A simple baseline metric of midbrain/inferior temporal ratio of flortaucipir uptake provided good to excellent differentiation between FTLD and high and low ADNC (sensitivities/specificities = 94%/95% and 71%/70%). INTERPRETATION There are cross-sectional and longitudinal differences in flortaucipir uptake between FTLD and high and low ADNC. However, optimum differentiation between FTLD and ADNC was achieved with baseline uptake rather than longitudinal rates. ANN NEUROL 2022;92:1016-1029.
Collapse
Affiliation(s)
| | | | | | | | - Farwa Ali
- Department of NeurologyMayo ClinicRochesterMNUSA
| | - Hugo Botha
- Department of NeurologyMayo ClinicRochesterMNUSA
| | | | - Mary M. Machulda
- Department of Psychiatry and PsychologyMayo ClinicRochesterMNUSA
| | | | | | - Matthew L. Senjem
- Department of RadiologyMayo ClinicRochesterMNUSA
- Department of Information TechnologyMayo ClinicRochesterMNUSA
| | | | | | | | | | - Julie A. Fields
- Department of Psychiatry and PsychologyMayo ClinicRochesterMNUSA
| | - Ross R. Reichard
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - Dennis W. Dickson
- Department of Neuroscience (Neurogenetics)Mayo ClinicJacksonvilleFLUSA
| | | | | | - Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMNUSA
| | | |
Collapse
|
17
|
Giannini LAA, Ohm DT, Rozemuller AJM, Dratch L, Suh E, van Deerlin VM, Trojanowski JQ, Lee EB, van Swieten JC, Grossman M, Seelaar H, Irwin DJ. Isoform-specific patterns of tau burden and neuronal degeneration in MAPT-associated frontotemporal lobar degeneration. Acta Neuropathol 2022; 144:1065-1084. [PMID: 36066634 PMCID: PMC9995405 DOI: 10.1007/s00401-022-02487-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/18/2022] [Accepted: 08/26/2022] [Indexed: 01/26/2023]
Abstract
Frontotemporal lobar degeneration with MAPT pathogenic variants (FTLD-MAPT) has heterogeneous tau pathological inclusions postmortem, consisting of three-repeat (3R) or four-repeat (4R) tau isoforms, or a combination (3R + 4R). Here, we studied grey matter tau burden, its relation to neuronal degeneration, and regional patterns of pathology in different isoform groups of FTLD-MAPT. We included 38 FTLD-MAPT autopsy cases with 10 different MAPT pathogenic variants, grouped based on predominant tau isoform(s). In up to eleven regions (ten cortical and one striatal), we quantified grey matter tau burden using digital histopathological analysis and assigned semi-quantitative ratings for neuronal degeneration (i.e. 0-4) and separate burden of glial and neuronal tau inclusions (i.e. 0-3). We used mixed modelling to compare pathology measures (1) across the entire cohort and (2) within isoform groups. In the total cohort, tau burden and neuronal degeneration were positively associated and most severe in the anterior temporal, anterior cingulate and transentorhinal cortices. Isoform groups showed distinctive features of tau burden and neuronal degeneration. Across all regions, the 3R isoform group had lower tau burden compared to the 4R group (p = 0.008), while at the same time showing more severe neuronal degeneration than the 4R group (p = 0.002). The 3R + 4R group had an intermediate profile with relatively high tau burden along with relatively severe neuronal degeneration. Neuronal tau inclusions were most frequent in the 4R group (p < 0.001 vs. 3R), while cortical glial tau inclusions were most frequent in the 3R + 4R and 4R groups (p ≤ 0.009 vs. 3R). Regionally, neuronal degeneration was consistently most severe in the anterior temporal cortex within each isoform group. In contrast, the regions with the highest tau burden differed in isoform groups (3R: striatum; 3R + 4R: striatum, inferior parietal lobule, middle frontal cortex, anterior cingulate cortex; 4R: transentorhinal cortex, anterior temporal cortex, fusiform gyrus). We conclude that FTLD-MAPT isoform groups show distinctive features of overall neuronal degeneration and regional tau burden, but all share pronounced anterior temporal neuronal degeneration. These data suggest that distinct isoform-related mechanisms of genetic tauopathies, with slightly divergent tau distribution, may share similar regional vulnerability to neurodegeneration within the frontotemporal paralimbic networks.
Collapse
Affiliation(s)
- Lucia A A Giannini
- Alzheimer Center, Department of Neurology, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Daniel T Ohm
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Frontotemporal Degeneration Center (FTDC), University of Pennsylvania Perelman School of Medicine, Hospital of the University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, The Netherlands
| | - Laynie Dratch
- Frontotemporal Degeneration Center (FTDC), University of Pennsylvania Perelman School of Medicine, Hospital of the University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - EunRan Suh
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vivianna M van Deerlin
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Edward B Lee
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John C van Swieten
- Alzheimer Center, Department of Neurology, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Murray Grossman
- Frontotemporal Degeneration Center (FTDC), University of Pennsylvania Perelman School of Medicine, Hospital of the University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - Harro Seelaar
- Alzheimer Center, Department of Neurology, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| | - David J Irwin
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Frontotemporal Degeneration Center (FTDC), University of Pennsylvania Perelman School of Medicine, Hospital of the University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
18
|
Wang X, Broce I, Deters KD, Fan CC, Banks SJ. Identification of Sex-Specific Genetic Variants Associated With Tau PET. Neurol Genet 2022; 8:e200043. [PMID: 36530928 PMCID: PMC9756308 DOI: 10.1212/nxg.0000000000200043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/22/2022] [Indexed: 12/13/2022]
Abstract
Background and Objectives Important sex differences exist in tau pathology along the Alzheimer disease (AD) continuum, with women showing enhanced tau deposition compared with men, especially during the mild cognitive impairment (MCI) phase. This study aims to identify specific genetic variants associated with sex differences in regional tau aggregation, as measured with PET. Methods Four hundred ninety-three participants (women, n = 246; men, n = 247) who self-identified as White from the AD Neuroimaging Initiative study, with genotyping data and 18F-Flortaucipir tau PET data, were included irrespective of clinical diagnosis (cognitively normal [CN], MCI, and AD). We focused on the genetic variants within 10 genes previously shown to have sex-dependent effects on AD to reduce the burden of multiple comparisons: BIN1, MS4A6A, DNAJA2, FERMT2, APOC1, APOC1P1, FAM193B, C2orf47, TYW5, and CR1. Multivariate analysis of variance was applied to identify genetic variants associated with tau PET data in 3 regions of interests (composite regions of Braak I, Braak III/IV, and Braak V/VI stages) in women and men separately. We controlled for age, scanner manufacture, amyloid status, APOE ε4 carriership, diagnosis (CN vs MCI vs AD), and the first 10 genetic principal components to adjust for population stratification. Results We identified 3 genetic loci within 3 different genes associated with tau deposits specifically in women: rs79711283 within DNAJA2, rs113357081 within FERMT2, and rs74614106 within TYW5. In men, we also identified 3 loci within CR1 associated with tau deposits: rs115096248, rs113698814, and rs78150633. Discussion Our findings revealed sex-specific genetic variants associated with tau deposition independent of APOE ε4, amyloid status, and clinical diagnosis. These results provide potential molecular targets for understanding the mechanism of sex-specific tau aggregation and developing sex-specific gene-guided precision prevention or therapeutic interventions for AD.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurosciences (X.W., I.B., K.D.D., C.C.F., S.J.B.), University of California; and Center for Multimodal Imaging and Genetics (X.W., I.B., C.C.F., S.J.B.), University of California, San Diego, La Jolla
| | - Iris Broce
- Department of Neurosciences (X.W., I.B., K.D.D., C.C.F., S.J.B.), University of California; and Center for Multimodal Imaging and Genetics (X.W., I.B., C.C.F., S.J.B.), University of California, San Diego, La Jolla
| | - Kacie D Deters
- Department of Neurosciences (X.W., I.B., K.D.D., C.C.F., S.J.B.), University of California; and Center for Multimodal Imaging and Genetics (X.W., I.B., C.C.F., S.J.B.), University of California, San Diego, La Jolla
| | - Chun Chieh Fan
- Department of Neurosciences (X.W., I.B., K.D.D., C.C.F., S.J.B.), University of California; and Center for Multimodal Imaging and Genetics (X.W., I.B., C.C.F., S.J.B.), University of California, San Diego, La Jolla
| | - Sarah Jane Banks
- Department of Neurosciences (X.W., I.B., K.D.D., C.C.F., S.J.B.), University of California; and Center for Multimodal Imaging and Genetics (X.W., I.B., C.C.F., S.J.B.), University of California, San Diego, La Jolla
| |
Collapse
|
19
|
Burkett BJ, Babcock JC, Lowe VJ, Graff-Radford J, Subramaniam RM, Johnson DR. PET Imaging of Dementia: Update 2022. Clin Nucl Med 2022; 47:763-773. [PMID: 35543643 DOI: 10.1097/rlu.0000000000004251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
ABSTRACT PET imaging plays an essential role in achieving earlier and more specific diagnoses of dementia syndromes, important for clinical prognostication and optimal medical management. This has become especially vital with the recent development of pathology-specific disease-modifying therapy for Alzheimer disease, which will continue to evolve and require methods to select appropriate treatment candidates. Techniques that began as research tools such as amyloid and tau PET have now entered clinical use, making nuclear medicine physicians and radiologists essential members of the care team. This review discusses recent changes in the understanding of dementia and examines the roles of nuclear medicine imaging in clinical practice. Within this framework, multiple cases will be shown to illustrate a systematic approach of FDG PET interpretation and integration of PET imaging of specific molecular pathology including dopamine transporters, amyloid, and tau. The approach presented here incorporates contemporary understanding of both common and uncommon dementia syndromes, intended as an updated practical guide to assist with the sophisticated interpretation of nuclear medicine examinations in the context of this rapidly and continually developing area of imaging.
Collapse
|
20
|
Krishnadas N, Doré V, Laws SM, Porter T, Lamb F, Bozinovski S, Villemagne VL, Rowe CC. Exploring discordant low amyloid beta and high neocortical tau positron emission tomography cases. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12326. [PMID: 36051174 PMCID: PMC9413469 DOI: 10.1002/dad2.12326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 11/12/2022]
Abstract
Introduction Neocortical 3R4R (3-repeat/4-repeat) tau aggregates are rarely observed in the absence of amyloid beta (Aβ). 18F-MK6240 binds specifically to the 3R4R form of tau that is characteristic of Alzheimer's disease (AD). We report four cases with negative Aβ, but positive tau positron emission tomography (PET) findings. Methods All Australian Imaging, Biomarkers and Lifestyle study of aging (AIBL) study participants with Aβ (18F-NAV4694) and tau (18F-MK6240) PET scans were included. Centiloid <25 defined negative Aβ PET (Aβ-). The presence of neocortical tau was defined quantitatively and visually. Results Aβ- PET was observed in 276 participants. Four of these participants (one cognitively unimpaired [CU], two mild cognitive impairment [MCI], one AD) had tau tracer retention in a pattern consistent with Braak tau stages V to VI. Fluid biomarkers supported a diagnosis of AD. In silico analysis of APP, PSEN1, PSEN2, and MAPT genes did not identify relevant functional mutations. Discussion Discordant cases were infrequent (1.4% of all Aβ- participants). In these cases, the Aβ PET ligand may not be detecting the Aβ that is present.
Collapse
Affiliation(s)
- Natasha Krishnadas
- Florey Department of Neurosciences & Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Department of Molecular Imaging & TherapyAustin HealthHeidelbergVictoriaAustralia
| | - Vincent Doré
- Department of Molecular Imaging & TherapyAustin HealthHeidelbergVictoriaAustralia
- Health and Biosecurity FlagshipThe Australian eHealth Research CentreMelbourneVictoriaAustralia
| | - Simon M. Laws
- Centre for Precision HealthEdith Cowan UniversityPerthWAAustralia
- Collaborative Genomics and Translation GroupSchool of Medical and Health SciencesEdith Cowan UniversityPerthWAAustralia
| | - Tenielle Porter
- Centre for Precision HealthEdith Cowan UniversityPerthWAAustralia
- Collaborative Genomics and Translation GroupSchool of Medical and Health SciencesEdith Cowan UniversityPerthWAAustralia
| | - Fiona Lamb
- Department of Molecular Imaging & TherapyAustin HealthHeidelbergVictoriaAustralia
| | - Svetlana Bozinovski
- Department of Molecular Imaging & TherapyAustin HealthHeidelbergVictoriaAustralia
| | - Victor L Villemagne
- Centre for Precision HealthEdith Cowan UniversityPerthWAAustralia
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Christopher C. Rowe
- Florey Department of Neurosciences & Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Department of Molecular Imaging & TherapyAustin HealthHeidelbergVictoriaAustralia
- Melbourne Dementia CenterFlorey Institute of Neuroscience & Mental HealthParkvilleVictoriaAustralia
| |
Collapse
|
21
|
Wang R, Gao H, Xie H, Jia Z, Chen Q. Molecular imaging biomarkers in familial frontotemporal lobar degeneration: Progress and prospects. Front Neurol 2022; 13:933217. [PMID: 36051222 PMCID: PMC9424494 DOI: 10.3389/fneur.2022.933217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/25/2022] [Indexed: 12/01/2022] Open
Abstract
Familial frontotemporal lobar degeneration (FTLD) is a pathologically heterogeneous group of neurodegenerative diseases with diverse genotypes and clinical phenotypes. Three major mutations were reported in patients with familial FTLD, namely, progranulin (GRN), microtubule-associated protein tau (MAPT), and the chromosome 9 open reading frame 72 (C9orf72) repeat expansion, which could cause neurodegenerative pathological changes years before symptom onset. Noninvasive quantitative molecular imaging with PET or single-photon emission CT (SPECT) allows for selective visualization of the molecular targets in vivo to investigate brain metabolism, perfusion, neuroinflammation, and pathophysiological changes. There was increasing evidence that several molecular imaging biomarkers tend to serve as biomarkers to reveal the early brain abnormalities in familial FTLD. Tau-PET with 18F-flortaucipir and 11C-PBB3 demonstrated the elevated tau position in patients with FTLD and also showed the ability to differentiate patterns among the different subtypes of the mutations in familial FTLD. Furthermore, dopamine transporter imaging with the 11C-DOPA and 11C-CFT in PET and the 123I-FP-CIT in SPECT revealed the loss of dopaminergic neurons in the asymptomatic and symptomatic patients of familial FTLD. In addition, PET imaging with the 11C-MP4A has demonstrated reduced acetylcholinesterase (AChE) activity in patients with FTLD, while PET with the 11C-DAA1106 and 11C-PK11195 revealed an increased level of microglial activation associated with neuroinflammation even before the onset of symptoms in familial FTLD. 18F-fluorodeoxyglucose (FDG)-PET indicated hypometabolism in FTLD with different mutations preceded the atrophy on MRI. Identifying molecular imaging biomarkers for familial FTLD is important for the in-vivo assessment of underlying pathophysiological changes with disease progression and future disease-modifying therapy. We review the recent progress of molecular imaging in familial FTLD with focused on the possible implication of these techniques and their prospects in specific mutation types.
Collapse
Affiliation(s)
- Ruihan Wang
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Hui Gao
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Hongsheng Xie
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Qin Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Qin Chen
| |
Collapse
|
22
|
Ruiz-Barrio I, Horta-Barba A, Illán-Gala I, Kulisevsky J, Pagonabarraga J. Genotype-Phenotype Correlation in Progressive Supranuclear Palsy Syndromes: Clinical and Radiological Similarities and Specificities. Front Neurol 2022; 13:861585. [PMID: 35557621 PMCID: PMC9087829 DOI: 10.3389/fneur.2022.861585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022] Open
Abstract
The progressive supranuclear palsy (PSP) syndrome encompasses different entities. PSP disease of sporadic origin is the most frequent presentation, but different genetic mutations can lead either to monogenic variants of PSP disease, or to other conditions with a different pathophysiology that eventually may result in PSP phenotype. PSP syndrome of monogenic origin is poorly understood due to the low prevalence and variable expressivity of some mutations. Through this review, we describe how early age of onset, family history of early dementia, parkinsonism, dystonia, or motor neuron disease among other clinical features, as well as some neuroimaging signatures, may be the important clues to suspect PSP syndrome of monogenic origin. In addition, a diagnostic algorithm is proposed that may be useful to guide the genetic diagnosis once there is clinical suspicion of a monogenic PSP syndrome.
Collapse
Affiliation(s)
- Iñigo Ruiz-Barrio
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ignacio Illán-Gala
- Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
23
|
Disclosing tau tangles using PET imaging: a pharmacological review of the radiotracers available in 2021. Acta Neurol Belg 2022; 122:263-272. [PMID: 34713414 DOI: 10.1007/s13760-021-01797-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/06/2021] [Indexed: 11/27/2022]
Abstract
Neurological symptoms depend on the topography of the lesions in the nervous system, hence the importance of brain imaging for neurologists. Neurological treatment, however, depends on the biological nature of the lesions. The development of radiotracers specific for the proteinopathies observed in neurodegenerative disorders is, therefore, crucially important for better understanding the relationships between the pathology and the clinical symptoms, as well as the efficacy of therapeutical interventions. The tau protein is involved in several neurodegenerative disorders, that can be distinguished both biologically and clinically as the type of tau isoforms and filaments observed in brain aggregates, and the brain regions affected differ between tauopathies. Over the past few years, several tracers have been developed for imaging tauopathies with positron emission tomography. The present review aims to compare the binding properties of these tracers, with a specific focus on how these properties might be relevant for neurologists using these biomarkers to characterize the pathology of patients presenting with clinical symptoms suspect of a neurodegenerative disorder.
Collapse
|
24
|
Kara F, Joers JM, Deelchand DK, Park YW, Przybelski SA, Lesnick TG, Senjem ML, Zeydan B, Knopman DS, Lowe VJ, Vemuri P, Mielke MM, Machulda MM, Jack CR, Petersen RC, Öz G, Kantarci K. 1H MR spectroscopy biomarkers of neuronal and synaptic function are associated with tau deposition in cognitively unimpaired older adults. Neurobiol Aging 2022; 112:16-26. [PMID: 35038671 PMCID: PMC8976711 DOI: 10.1016/j.neurobiolaging.2021.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 12/25/2022]
Abstract
Proton magnetic resonance spectroscopy (1H MRS) may provide information on pathophysiological changes associated with tau deposition in cognitively unimpaired older adults. In this study, the associations of posterior cingulate gyrus tau and amyloid beta (Aβ) deposition on PET with 1H MRS metabolite ratios acquired from bilateral posterior cingulate gyri were investigated in cognitively unimpaired older adults. Participants (n = 40) from the Mayo Clinic Study of Aging underwent single-voxel sLASER 1H MRS from the posterior cingulate gyrus at 3 Tesla, 18F-flortaucipir, and 11C- Pittsburgh Compound B (PiB) PET. An increase in posterior cingulate gyrus tau deposition, but not elevated Aβ, was associated with lower N-acetylaspartate/total creatine (tCr) and glutamate (Glu)/tCr ratios, and sex by tau interaction was observed in association with Glu/tCr. Higher tau levels in cognitively unimpaired older adults are associated with biomarkers of neural and synaptic injury even in the absence of cognitive impairment and these relationships appear to be stronger in women than in men.
Collapse
Affiliation(s)
- Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - James M Joers
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Dinesh K Deelchand
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Young Woo Park
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Burcu Zeydan
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | | | | | - Gülin Öz
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
25
|
Benatar M, Wuu J, McHutchison C, Postuma RB, Boeve BF, Petersen R, Ross CA, Rosen H, Arias JJ, Fradette S, McDermott MP, Shefner J, Stanislaw C, Abrahams S, Cosentino S, Andersen PM, Finkel RS, Granit V, Grignon AL, Rohrer JD, McMillan CT, Grossman M, Al-Chalabi A, Turner MR. Preventing amyotrophic lateral sclerosis: insights from pre-symptomatic neurodegenerative diseases. Brain 2022; 145:27-44. [PMID: 34677606 PMCID: PMC8967095 DOI: 10.1093/brain/awab404] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/16/2021] [Accepted: 10/08/2021] [Indexed: 11/12/2022] Open
Abstract
Significant progress has been made in understanding the pre-symptomatic phase of amyotrophic lateral sclerosis. While much is still unknown, advances in other neurodegenerative diseases offer valuable insights. Indeed, it is increasingly clear that the well-recognized clinical syndromes of Alzheimer's disease, Parkinson's disease, Huntington's disease, spinal muscular atrophy and frontotemporal dementia are also each preceded by a pre-symptomatic or prodromal period of varying duration, during which the underlying disease process unfolds, with associated compensatory changes and loss of inherent system redundancy. Key insights from these diseases highlight opportunities for discovery in amyotrophic lateral sclerosis. The development of biomarkers reflecting amyloid and tau has led to a shift in defining Alzheimer's disease based on inferred underlying histopathology. Parkinson's disease is unique among neurodegenerative diseases in the number and diversity of non-genetic biomarkers of pre-symptomatic disease, most notably REM sleep behaviour disorder. Huntington's disease benefits from an ability to predict the likely timing of clinically manifest disease based on age and CAG-repeat length alongside reliable neuroimaging markers of atrophy. Spinal muscular atrophy clinical trials have highlighted the transformational value of early therapeutic intervention, and studies in frontotemporal dementia illustrate the differential role of biomarkers based on genotype. Similar advances in amyotrophic lateral sclerosis would transform our understanding of key events in pathogenesis, thereby dramatically accelerating progress towards disease prevention. Deciphering the biology of pre-symptomatic amyotrophic lateral sclerosis relies on a clear conceptual framework for defining the earliest stages of disease. Clinically manifest amyotrophic lateral sclerosis may emerge abruptly, especially among those who harbour genetic mutations associated with rapidly progressive amyotrophic lateral sclerosis. However, the disease may also evolve more gradually, revealing a prodromal period of mild motor impairment preceding phenoconversion to clinically manifest disease. Similarly, cognitive and behavioural impairment, when present, may emerge gradually, evolving through a prodromal period of mild cognitive impairment or mild behavioural impairment before progression to amyotrophic lateral sclerosis. Biomarkers are critically important to studying pre-symptomatic amyotrophic lateral sclerosis and essential to efforts to intervene therapeutically before clinically manifest disease emerges. The use of non-genetic biomarkers, however, presents challenges related to counselling, informed consent, communication of results and limited protections afforded by existing legislation. Experiences from pre-symptomatic genetic testing and counselling, and the legal protections against discrimination based on genetic data, may serve as a guide. Building on what we have learned-more broadly from other pre-symptomatic neurodegenerative diseases and specifically from amyotrophic lateral sclerosis gene mutation carriers-we present a road map to early intervention, and perhaps even disease prevention, for all forms of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Michael Benatar
- Department of Neurology, University of Miami, Miami, FL, USA
| | - Joanne Wuu
- Department of Neurology, University of Miami, Miami, FL, USA
| | - Caroline McHutchison
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Ronald B Postuma
- Department of Neurology, Montreal Neurological Institute, McGill University, Montreal, Canada
| | | | | | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Howard Rosen
- Department of Neurology, University of California San Francisco, CA, USA
| | - Jalayne J Arias
- Department of Neurology, University of California San Francisco, CA, USA
| | | | - Michael P McDermott
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jeremy Shefner
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Sharon Abrahams
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | | | - Peter M Andersen
- Department of Clinical Science, Neurosciences, Umeå University, Sweden
| | - Richard S Finkel
- Department of Pediatric Medicine, Center for Experimental Neurotherapeutics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Volkan Granit
- Department of Neurology, University of Miami, Miami, FL, USA
| | | | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - Corey T McMillan
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, UK
- Department of Neurology, King's College Hospital, London, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Silva MC, Nandi G, Donovan KA, Cai Q, Berry BC, Nowak RP, Fischer ES, Gray NS, Ferguson FM, Haggarty SJ. Discovery and Optimization of Tau Targeted Protein Degraders Enabled by Patient Induced Pluripotent Stem Cells-Derived Neuronal Models of Tauopathy. Front Cell Neurosci 2022; 16:801179. [PMID: 35317195 PMCID: PMC8934437 DOI: 10.3389/fncel.2022.801179] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/26/2022] [Indexed: 12/21/2022] Open
Abstract
Accumulation of misfolded, aggregating proteins concurrent with disease onset and progression is a hallmark of neurodegenerative proteinopathies. An important class of these are tauopathies, such as frontotemporal dementia (FTD) and Alzheimer’s disease (AD), associated with accumulation of aberrant forms of tau protein in the brain. Pathological tau undergoes abnormal post-translational modifications, misfolding, oligomerization and changes in solubility, cellular redistribution, and spreading. Development and testing of experimental therapeutics that target these pathological tau conformers requires use of cellular models that recapitulate neuronal endogenous, non-heterologous tau expression under genomic and physiological contexts relevant to disease. In this study, we employed FTD-patient induced pluripotent stem cells (iPSC)-derived neurons, expressing a tau variant or mutation, as primary models for driving a medicinal chemistry campaign around tau targeting degrader series. Our screening goal was to establish structure-activity relationships (SAR) for the different chemical series to identify the molecular composition that most efficiently led to tau degradation in human FTD ex vivo neurons. We describe the identification of the lead compound QC-01-175 and follow-up optimization strategies for this molecule. We present three final lead molecules with tau degradation activity in mutant neurons, which establishes potential disease relevance and will drive future studies on specificity and pharmacological properties.
Collapse
Affiliation(s)
- M. Catarina Silva
- Chemical Neurobiology Laboratory, Department of Neurology and Psychiatry, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Ghata Nandi
- Chemical Neurobiology Laboratory, Department of Neurology and Psychiatry, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Katherine A. Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Quan Cai
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Bethany C. Berry
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Radoslaw P. Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Nathanael S. Gray
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Fleur M. Ferguson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
- *Correspondence: Fleur M. Ferguson,
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Department of Neurology and Psychiatry, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Stephen J. Haggarty,
| |
Collapse
|
27
|
Boeve BF, Boxer AL, Kumfor F, Pijnenburg Y, Rohrer JD. Advances and controversies in frontotemporal dementia: diagnosis, biomarkers, and therapeutic considerations. Lancet Neurol 2022; 21:258-272. [DOI: 10.1016/s1474-4422(21)00341-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 08/16/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022]
|
28
|
Benussi A, Alberici A, Samra K, Russell LL, Greaves CV, Bocchetta M, Ducharme S, Finger E, Fumagalli G, Galimberti D, Jiskoot LC, Le Ber I, Masellis M, Nacmias B, Rowe JB, Sanchez-Valle R, Seelaar H, Synofzik M, Rohrer JD, Borroni B. Conceptual framework for the definition of preclinical and prodromal frontotemporal dementia. Alzheimers Dement 2021; 18:1408-1423. [PMID: 34874596 DOI: 10.1002/alz.12485] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022]
Abstract
The presymptomatic stages of frontotemporal dementia (FTD) are still poorly defined and encompass a long accrual of progressive biological (preclinical) and then clinical (prodromal) changes, antedating the onset of dementia. The heterogeneity of clinical presentations and the different neuropathological phenotypes have prevented a prior clear description of either preclinical or prodromal FTD. Recent advances in therapeutic approaches, at least in monogenic disease, demand a proper definition of these predementia stages. It has become clear that a consensus lexicon is needed to comprehensively describe the stages that anticipate dementia. The goal of the present work is to review existing literature on the preclinical and prodromal phases of FTD, providing recommendations to address the unmet questions, therefore laying out a strategy for operationalizing and better characterizing these presymptomatic disease stages.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy
| | - Antonella Alberici
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy
| | - Kiran Samra
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Caroline V Greaves
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Simon Ducharme
- Department of Psychiatry, Douglas Mental Health University Institute and Douglas Research Centre, McGill University, Montreal, Québec, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Giorgio Fumagalli
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy.,University of Milan, Milan, Italy
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy.,University of Milan, Milan, Italy
| | - Lize C Jiskoot
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Department of Neurology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Isabelle Le Ber
- Paris Brain Institute - Institut du Cerveau - ICM, Sorbonne Université, Inserm U1127, CNRS UMR, Paris, France.,Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Reference Network for Rare Neurological Diseases (ERN-RND), Paris, France
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, and IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, MRC Cognition and Brain Sciences Unit and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
29
|
Ricci M, Cimini A, Camedda R, Chiaravalloti A, Schillaci O. Tau Biomarkers in Dementia: Positron Emission Tomography Radiopharmaceuticals in Tauopathy Assessment and Future Perspective. Int J Mol Sci 2021; 22:ijms222313002. [PMID: 34884804 PMCID: PMC8657996 DOI: 10.3390/ijms222313002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/14/2021] [Accepted: 11/25/2021] [Indexed: 01/20/2023] Open
Abstract
Abnormal accumulation of Tau protein is closely associated with neurodegeneration and cognitive impairment and it is a biomarker of neurodegeneration in the dementia field, especially in Alzheimer’s disease (AD); therefore, it is crucial to be able to assess the Tau deposits in vivo. Beyond the fluid biomarkers of tauopathy described in this review in relationship with the brain glucose metabolic patterns, this review aims to focus on tauopathy assessment by using Tau PET imaging. In recent years, several first-generation Tau PET tracers have been developed and applied in the dementia field. Common limitations of first-generation tracers include off-target binding and subcortical white-matter uptake; therefore, several institutions are working on developing second-generation Tau tracers. The increasing knowledge about the distribution of first- and second-generation Tau PET tracers in the brain may support physicians with Tau PET data interpretation, both in the research and in the clinical field, but an updated description of differences in distribution patterns among different Tau tracers, and in different clinical conditions, has not been reported yet. We provide an overview of first- and second-generation tracers used in ongoing clinical trials, also describing the differences and the properties of novel tracers, with a special focus on the distribution patterns of different Tau tracers. We also describe the distribution patterns of Tau tracers in AD, in atypical AD, and further neurodegenerative diseases in the dementia field.
Collapse
Affiliation(s)
- Maria Ricci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
- Correspondence:
| | - Andrea Cimini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
| | - Riccardo Camedda
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (A.C.); (R.C.); (A.C.); (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
30
|
Zhou XY, Lu JY, Liu FT, Wu P, Zhao J, Ju ZZ, Tang YL, Shi QY, Lin HM, Wu JJ, Yen TC, Zuo CT, Sun YM, Wang J. In Vivo 18 F-APN-1607 Tau Positron Emission Tomography Imaging in MAPT Mutations: Cross-Sectional and Longitudinal Findings. Mov Disord 2021; 37:525-534. [PMID: 34842301 DOI: 10.1002/mds.28867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/01/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Frontotemporal lobar degeneration with tauopathy caused by MAPT (microtubule-associated protein tau) mutations is a highly heterogenous disorder. The ability to visualize and longitudinally monitor tau deposits may be beneficial to understand disease pathophysiology and predict clinical trajectories. OBJECTIVE The aim of this study was to investigate the cross-sectional and longitudinal 18 F-APN-1607 positron emission tomography/computed tomography (PET/CT) imaging findings in MAPT mutation carriers. METHODS Seven carriers of MAPT mutations (six within exon 10 and one outside of exon 10) and 15 healthy control subjects were included. All participants underwent 18 F-APN-1607 PET/CT at baseline. Three carriers of exon 10 mutations received follow-up 18 F-APN-1607 PET/CT scans. Standardized uptake value ratio (SUVR) maps were obtained using the cerebellar gray matter as the reference region. SUVR values observed in MAPT mutation carriers were normalized to data from healthy control subjects. A regional SUVR z score ≥ 2 was used as the criterion to define positive 18 F-APN-1607 PET/CT findings. RESULTS Although the seven study patients had heterogenous clinical phenotypes, all showed a significant 18 F-APN-1607 uptake characterized by high-contrast signals. However, the anatomical localization of tau deposits differed in patients with distinct clinical symptoms. Follow-up imaging data, which were available for three patients, demonstrated worsening trends in patterns of tau accumulation over time, which were paralleled by a significant clinical deterioration. CONCLUSIONS Our data represent a promising step in understanding the usefulness of 18 F-APN-1607 PET/CT imaging for detecting tau accumulation in MAPT mutation carriers. Our preliminary follow-up data also suggest the potential value of 18 F-APN-1607 PET/CT for monitoring the longitudinal trajectories of frontotemporal lobar degeneration caused by MAPT mutations. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xin-Yue Zhou
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia-Ying Lu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Feng-Tao Liu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jue Zhao
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zi-Zhao Ju
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Lin Tang
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing-Yi Shi
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Hua-Mei Lin
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian-Jun Wu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Min Sun
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Schwarz CG, Knopman DS, Ramanan VK, Lowe VJ, Wiste HJ, Cogswell PM, Utianski RL, Senjem ML, Gunter JR, Vemuri P, Petersen RC, Jack CR. Longitudinally Increasing Elevated Asymmetric Flortaucipir Binding in a Cognitively Unimpaired Amyloid-Negative Older Individual. J Alzheimers Dis 2021; 85:59-64. [PMID: 34776445 PMCID: PMC8842786 DOI: 10.3233/jad-215052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We present the case of a cognitively unimpaired 77-year-old man with elevated, asymmetric, and longitudinally increasing Flortaucipir tau PET despite normal (visually negative) amyloid PET. His atypical tau PET signal persisted and globally increased in a follow-up scan five years later. Across eight years of observations, temporoparietal atrophy was observed consistent with tau PET patterns, but he retained the cognitively unimpaired classification. Altogether, his atypical tau PET signal is not explained by any known risk factors or alternative pathologies, and other imaging findings were not remarkable. He remains enrolled for further observation.
Collapse
Affiliation(s)
| | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.,Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
32
|
Song M, Beyer L, Kaiser L, Barthel H, van Eimeren T, Marek K, Nitschmann A, Scheifele M, Palleis C, Respondek G, Kern M, Biechele G, Hammes J, Bischof G, Barbe M, Onur Ö, Jessen F, Saur D, Schroeter ML, Rumpf JJ, Rullmann M, Schildan A, Patt M, Neumaier B, Barret O, Madonia J, Russell DS, Stephens AW, Mueller A, Roeber S, Herms J, Bötzel K, Danek A, Levin J, Classen J, Höglinger GU, Bartenstein P, Villemagne V, Drzezga A, Seibyl J, Sabri O, Boening G, Ziegler S, Brendel M. Binding characteristics of [ 18F]PI-2620 distinguish the clinically predicted tau isoform in different tauopathies by PET. J Cereb Blood Flow Metab 2021; 41:2957-2972. [PMID: 34044665 PMCID: PMC8545042 DOI: 10.1177/0271678x211018904] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The novel tau-PET tracer [18F]PI-2620 detects the 3/4-repeat-(R)-tauopathy Alzheimer's disease (AD) and the 4R-tauopathies corticobasal syndrome (CBS) and progressive supranuclear palsy (PSP). We determined whether [18F]PI-2620 binding characteristics deriving from non-invasive reference tissue modelling differentiate 3/4R- and 4R-tauopathies. Ten patients with a 3/4R tauopathy (AD continuum) and 29 patients with a 4R tauopathy (CBS, PSP) were evaluated. [18F]PI-2620 PET scans were acquired 0-60 min p.i. and the distribution volume ratio (DVR) was calculated. [18F]PI-2620-positive clusters (DVR ≥ 2.5 SD vs. 11 healthy controls) were evaluated by non-invasive kinetic modelling. R1 (delivery), k2 & k2a (efflux), DVR, 30-60 min standardized-uptake-value-ratios (SUVR30-60) and the linear slope of post-perfusion phase SUVR (9-60 min p.i.) were compared between 3/4R- and 4R-tauopathies. Cortical clusters of 4R-tau cases indicated higher delivery (R1SRTM: 0.92 ± 0.21 vs. 0.83 ± 0.10, p = 0.0007), higher efflux (k2SRTM: 0.17/min ±0.21/min vs. 0.06/min ± 0.07/min, p < 0.0001), lower DVR (1.1 ± 0.1 vs. 1.4 ± 0.2, p < 0.0001), lower SUVR30-60 (1.3 ± 0.2 vs. 1.8 ± 0.3, p < 0.0001) and flatter slopes of the post-perfusion phase (slope9-60: 0.006/min ± 0.007/min vs. 0.016/min ± 0.008/min, p < 0.0001) when compared to 3/4R-tau cases. [18F]PI-2620 binding characteristics in cortical regions differentiate 3/4R- and 4R-tauopathies. Higher tracer clearance indicates less stable binding in 4R tauopathies when compared to 3/4R-tauopathies.
Collapse
Affiliation(s)
- Mengmeng Song
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Thilo van Eimeren
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany.,Department of Neurology, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ken Marek
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - Alexander Nitschmann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Gesine Respondek
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Maike Kern
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Hammes
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Gèrard Bischof
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Michael Barbe
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Özgür Onur
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry, University Hospital Cologne, Cologne, Germany.,Center for Memory Disorders, University Hospital Cologne, Cologne, Germany
| | - Dorothee Saur
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University of Leipzig, Leipzig, Germany.,LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.,Max- Planck-Institute of Human Cognitive and Brain Sciences, Leipzig, Germany.,FTLD Consortium Germany, Ulm, Germany
| | | | - Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Andreas Schildan
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Bernd Neumaier
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Olivier Barret
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA.,Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, CEA, CNRS, MIRCen, Fontenay-aux-Roses, France
| | - Jennifer Madonia
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - David S Russell
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | | | | | - Sigrun Roeber
- Center for Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Center for Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Kai Bötzel
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrian Danek
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Günter U Höglinger
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Technical University Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Victor Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - John Seibyl
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Guido Boening
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
33
|
Levy JP, Bezgin G, Savard M, Pascoal TA, Finger E, Laforce R, Sonnen JA, Soucy JP, Gauthier S, Rosa-Neto P, Ducharme S. 18F-MK-6240 tau-PET in genetic frontotemporal dementia. Brain 2021; 145:1763-1772. [PMID: 34664612 PMCID: PMC9166561 DOI: 10.1093/brain/awab392] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/25/2021] [Accepted: 09/10/2021] [Indexed: 11/28/2022] Open
Abstract
Tau is one of several proteins associated with frontotemporal dementia. While knowing which protein is causing a patient’s disease is crucial, no biomarker currently exists for identifying tau in vivo in frontotemporal dementia. The objective of this study was to investigate the potential for the promising 18F-MK-6240 PET tracer to bind to tau in vivo in genetic frontotemporal dementia. We enrolled subjects with genetic frontotemporal dementia, who constitute an ideal population for testing because their pathology is already known based on their mutation. Ten participants (three with symptomatic P301L and R406W MAPT mutations expected to show tau binding, three with presymptomatic MAPT mutations and four with non-tau mutations who acted as disease controls) underwent clinical characterization, tau-PET scanning with 18F-MK-6240, amyloid-PET imaging with 18F-NAV-4694 to rule out confounding Alzheimer’s pathology, and high-resolution structural MRI. Tau-PET scans of all three symptomatic MAPT carriers demonstrated at least mild 18F-MK-6240 binding in expected regions, with particularly strong binding in a subject with an R406W MAPT mutation (known to be associated with Alzheimer’s like neurofibrillary tangles). Two asymptomatic MAPT carriers estimated to be 5 years from disease onset both showed modest 18F-MK-6240 binding, while one ∼30 years from disease onset did not exhibit any binding. Additionally, four individuals with symptomatic frontotemporal dementia caused by a non-tau mutation were scanned (two C9orf72; one GRN; one VCP): 18F-MK-6240 scans were negative for three subjects, while one advanced C9orf72 case showed minimal regionally non-specific binding. All 10 amyloid-PET scans were negative. Furthermore, a general linear model contrasting genetic frontotemporal dementia subjects to a set of 83 age-matched controls showed significant binding only in the MAPT carriers in selected frontal, temporal and subcortical regions. In summary, our findings demonstrate mild but significant binding of MK-6240 in amyloid-negative P301L and R406W MAPT mutation subjects, with higher standardized uptake value ratio in the R406W mutation associated with the presence of NFTs, and little non-specific binding. These results highlight that a positive 18F-MK-6240 tau-PET does not necessarily imply a diagnosis of Alzheimer’s disease and point towards a potential use for 18F-MK-6240 as a biomarker in certain tauopathies beyond Alzheimer’s, although further patient recruitment and autopsy studies will be necessary to determine clinical applicability.
Collapse
Affiliation(s)
- Jake P Levy
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Parkwood Institute, Lawson Health Research Institute, University of Western Ontario, London, ON, Canada
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques du CHU de Québec, Faculté de Médecine, Université Laval, QC, Canada
| | - Joshua A Sonnen
- Departments of Pathology, Neurology and Neurosurgery, Montreal Neurological Institute, McGill University
| | - Jean-Paul Soucy
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Pedro Rosa-Neto
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada.,Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada.,Douglas Mental Health University Institute, Department of Psychiatry, Montreal, QC H4H 1R3, Canada
| |
Collapse
|
34
|
Wolters EE, Papma JM, Verfaillie SCJ, Visser D, Weltings E, Groot C, van der Ende EL, Giannini LAA, Tuncel H, Timmers T, Boellaard R, Yaqub M, van Assema DME, Kuijper DA, Segbers M, Rozemuller AJM, Barkhof F, Windhorst AD, van der Flier WM, Pijnenburg YAL, Scheltens P, van Berckel BNM, van Swieten JC, Ossenkoppele R, Seelaar H. [ 18F]Flortaucipir PET Across Various MAPT Mutations in Presymptomatic and Symptomatic Carriers. Neurology 2021; 97:e1017-e1030. [PMID: 34210823 PMCID: PMC8448551 DOI: 10.1212/wnl.0000000000012448] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/07/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the [18F]flortaucipir binding distribution across MAPT mutations in presymptomatic and symptomatic carriers. METHODS We compared regional [18F]flortaucipir binding potential (BPND) derived from a 130-minute dynamic [18F]flortaucipir PET scan in 9 (pre)symptomatic MAPT mutation carriers (4 with P301L [1 symptomatic], 2 with R406W [1 symptomatic], 1 presymptomatic L315R, 1 presymptomatic S320F, and 1 symptomatic G272V carrier) with 30 cognitively normal controls and 52 patients with Alzheimer disease. RESULTS [18F]Flortaucipir BPND images showed overall highest binding in the symptomatic carriers. This was most pronounced in the symptomatic R406W carrier in whom tau binding exceeded the normal control range in the anterior cingulate cortex, insula, amygdala, temporal, parietal, and frontal lobe. Elevated medial temporal lobe BPND was observed in a presymptomatic R406W carrier. The single symptomatic carrier and 1 of the 3 presymptomatic P301L carriers showed elevated [18F]flortaucipir BPND in the insula, parietal, and frontal lobe compared to controls. The symptomatic G272V carrier exhibited a widespread elevated cortical BPND, with at neuropathologic examination a combination of 3R pathology and encephalitis. The L315R presymptomatic mutation carrier showed higher frontal BPND compared to controls. The BPND values of the S320F presymptomatic mutation carrier fell within the range of controls. CONCLUSION Presymptomatic MAPT mutation carriers already showed subtle elevated tau binding, whereas symptomatic MAPT mutation carriers showed a more marked increase in [18F]flortaucipir BPND. Tau deposition was most pronounced in R406W MAPT (pre)symptomatic mutation carriers, which is associated with both 3R and 4R tau accumulation. Thus, [18F]flortaucipir may serve as an early biomarker for MAPT mutation carriers in mutations that cause 3R/4R tauopathies.
Collapse
Affiliation(s)
- Emma E Wolters
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden.
| | - Janne M Papma
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Sander C J Verfaillie
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Denise Visser
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Emma Weltings
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Colin Groot
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Emma L van der Ende
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Lucia A A Giannini
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Hayel Tuncel
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Tessa Timmers
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Ronald Boellaard
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Maqsood Yaqub
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Danielle M E van Assema
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Dennis A Kuijper
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Marcel Segbers
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Annemieke J M Rozemuller
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Frederik Barkhof
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Albert D Windhorst
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Wiesje M van der Flier
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Yolande A L Pijnenburg
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Philip Scheltens
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Bart N M van Berckel
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - John C van Swieten
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Rik Ossenkoppele
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Harro Seelaar
- From the Department of Radiology & Nuclear Medicine (E.E.W., S.C.J.V., D.V., E.W., H.T., T.T., R.B., M.Y., F.B., A.D.W., B.N.M.v.B.) and Alzheimer Center Amsterdam, Department of Neurology (E.E.W., C.G., W.M.v.d.F., Y.A.L.P., P.S., R.O.), Amsterdam Neuroscience, and Department of Epidemiology and Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC; Department of Neurology, Alzheimer Center (J.M.P., E.L.v.d.E., L.A.A.G., J.C.v.S., H.S.), and Department of Radiology & Nuclear Medicine (D.M.E.v.A., D.A.K., M.S.), Erasmus MC University Medical Center, Rotterdam; Department of Pathology (A.J.M.R.), Amsterdam Neuroscience, Amsterdam UMC, location VUmc, the Netherlands; Institutes of Neurology & Healthcare Engineering (F.B.), UCL, London, UK; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| |
Collapse
|
35
|
Sato C, Mallipeddi N, Ghoshal N, Wright BA, Day GS, Davis AA, Kim AH, Zipfel GJ, Bateman RJ, Gabelle A, Barthélemy NR. MAPT R406W increases tau T217 phosphorylation in absence of amyloid pathology. Ann Clin Transl Neurol 2021; 8:1817-1830. [PMID: 34342183 PMCID: PMC8419397 DOI: 10.1002/acn3.51435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 01/07/2023] Open
Abstract
Objective Tau hyperphosphorylation at threonine 217 (pT217) in cerebrospinal fluid (CSF) has recently been linked to early amyloidosis and could serve as a highly sensitive biomarker for Alzheimer’s disease (AD). However, it remains unclear whether other tauopathies induce pT217 modifications. To determine if pT217 modification is specific to AD, CSF pT217 was measured in AD and other tauopathies. Methods Using immunoprecipitation and mass spectrometry methods, we compared CSF T217 phosphorylation occupancy (pT217/T217) and amyloid‐beta (Aβ) 42/40 ratio in cognitively normal individuals and those with symptomatic AD, progressive supranuclear palsy, corticobasal syndrome, and sporadic and familial frontotemporal dementia. Results Individuals with AD had high CSF pT217/T217 and low Aβ42/40. In contrast, cognitively normal individuals and the majority of those with 4R tauopathies had low CSF pT217/T217 and normal Aβ 42/40. We identified a subgroup of individuals with increased CSF pT217/T217 and normal Aβ 42/40 ratio, most of whom were MAPT R406W mutation carriers. Diagnostic accuracies of CSF Aβ 42/40 and CSF pT217/T217, alone and in combination were compared. We show that CSF pT217/T217 × CSF Aβ 42/40 is a sensitive composite biomarker that can separate MAPT R406W carriers from cognitively normal individuals and those with other tauopathies. Interpretation MAPT R406W is a tau mutation that leads to 3R+4R tauopathy similar to AD, but without amyloid neuropathology. These findings suggest that change in CSF pT217/T217 ratio is not specific to AD and might reflect common downstream tau pathophysiology common to 3R+4R tauopathies.
Collapse
Affiliation(s)
- Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Nipun Mallipeddi
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Nupur Ghoshal
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Brenton A Wright
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, California
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida
| | - Albert A Davis
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri
| | - Albert H Kim
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri.,Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gregory J Zipfel
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri.,Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri.,Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Audrey Gabelle
- Department of Neurology, Memory Research and Resources Center, University Hospital of Montpellier, Neurosciences Institute of Montpellier, University of Montpellier, Montpellier, France
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
36
|
Su Y, Fu J, Yu J, Zhao Q, Guan Y, Zuo C, Li M, Tan H, Cheng X. Tau PET Imaging with [18F]PM-PBB3 in Frontotemporal Dementia with MAPT Mutation. J Alzheimers Dis 2021; 76:149-157. [PMID: 32444551 DOI: 10.3233/jad-200287] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Flortaucipir (AV-1451) and pyridinyl-butadienyl-benzothiazole 3 (PBB3) are newly developed and commonly used positron emission tomography (PET) tracers to detect tau deposition in tauopathies, including frontotemporal dementia (FTD). [18F]PM-PBB3, as a second-generation compound, has not been described in FTD so far. OBJECTIVE We aim to explore the in vivo performance of [18F]PM-PBB3 tau PET in an FTD case caused by microtubule-associated protein tau (MAPT) mutation and compare the binding to different tau strains between AV-1451 and PBB3. METHODS We reported the clinical and FDG, [18F]AV45 amyloid and [18F]PM-PBB3 tau PET findings in a patient with FTD of P301L MAPT mutation. Based on our results and published data, we summarized and compared the different utilities of tau PET tracers of AV-1451 and PBB3 in FTD with MAPT mutation. RESULTS The patient demonstrated slightly diffuse [18F]PM-PBB3 tau deposition in cerebral lobes especially in the left frontal lobe overlapping with the hypometabolic region detected by FDG PET. From our analysis of 35 FTD patients with MAPT mutation who underwent tau PET, AV-1451 was positive in all (n = 11) patients with mutations known to cause three and four repeat (3R/4R) tau deposition and in 14.3% (n = 2/14) of 4R tauopathies, while positive PBB3 retention was found in all patients with both 3R/4R (n = 2) and 4R (n = 8) tau. CONCLUSIONS [18F]PM-PBB3 tau PET assisted the diagnosis of FTD with P301L MAPT mutation, and might be useful in the in vivo detection of both 3R/4R and 4R tau domains in the brain of FTD with MAPT mutation.
Collapse
Affiliation(s)
- Ya Su
- Department of Neurology, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Jiayu Fu
- Department of Neurology, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Jintai Yu
- Department of Neurology, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Qianhua Zhao
- Department of Neurology, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Centre, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Centre, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming Li
- PET Centre, Huashan Hospital, Fudan University, Shanghai, China
| | - Haibo Tan
- PET Centre, Huashan Hospital, Fudan University, Shanghai, China
| | - Xin Cheng
- Department of Neurology, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Pascual B, Funk Q, Zanotti-Fregonara P, Cykowski MD, Veronese M, Rockers E, Bradbury K, Yu M, Nakawah MO, Román GC, Schulz PE, Arumanayagam AS, Beers D, Faridar A, Fujita M, Appel SH, Masdeu JC. Neuroinflammation is highest in areas of disease progression in semantic dementia. Brain 2021; 144:1565-1575. [PMID: 33824991 DOI: 10.1093/brain/awab057] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/25/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Despite epidemiological and genetic data linking semantic dementia to inflammation, the topography of neuroinflammation in semantic dementia, also known as the semantic variant of primary progressive aphasia, remains unclear. The pathology starts at the tip of the left temporal lobe where, in addition to cortical atrophy, a strong signal appears with the tau PET tracer 18F-flortaucipir, even though the disease is not typically associated with tau but with TDP-43 protein aggregates. Here, we characterized the topography of inflammation in semantic variant primary progressive aphasia using high-resolution PET and the tracer 11C-PBR28 as a marker of microglial activation. We also tested the hypothesis that inflammation, by providing non-specific binding targets, could explain the 18F-flortaucipir signal in semantic variant primary progressive aphasia. Eight amyloid-PET-negative patients with semantic variant primary progressive aphasia underwent 11C-PBR28 and 18F-flortaucipir PET. Healthy controls underwent 11C-PBR28 PET (n = 12) or 18F-flortaucipir PET (n = 12). Inflammation in PET with 11C-PBR28 was analysed using Logan graphical analysis with a metabolite-corrected arterial input function. 18F-flortaucipir standardized uptake value ratios were calculated using the cerebellum as the reference region. Since monoamine oxidase B receptors are expressed by astrocytes in affected tissue, selegiline was administered to one patient with semantic variant primary progressive aphasia before repeating 18F-flortaucipir scanning to test whether monoamine oxidase B inhibition blocked flortaucipir binding, which it did not. While 11C-PBR28 uptake was mostly cortical, 18F-flortaucipir uptake was greatest in the white matter. The uptake of both tracers was increased in the left temporal lobe and in the right temporal pole, as well as in regions adjoining the left temporal pole such as insula and orbitofrontal cortex. However, peak uptake of 18F-flortaucipir localized to the left temporal pole, the epicentre of pathology, while the peak of inflammation 11C-PBR28 uptake localized to a more posterior, mid-temporal region and left insula and orbitofrontal cortex, in the periphery of the damage core. Neuroinflammation, greatest in the areas of progression of the pathological process in semantic variant primary progressive aphasia, should be further studied as a possible therapeutic target to slow disease progression.
Collapse
Affiliation(s)
- Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Quentin Funk
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA.,Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, King's College London, London, UK
| | - Elijah Rockers
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Kathleen Bradbury
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Meixiang Yu
- Cyclotron and Radiopharmaceutical Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Mohammad O Nakawah
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Gustavo C Román
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Paul E Schulz
- Department of Neurology, McGovern Medical School of UT Health, Houston, TX, USA
| | - Anithachristy S Arumanayagam
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - David Beers
- Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Alireza Faridar
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Masahiro Fujita
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Stanley H Appel
- Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX, USA
| |
Collapse
|
38
|
Peet BT, Spina S, Mundada N, La Joie R. Neuroimaging in Frontotemporal Dementia: Heterogeneity and Relationships with Underlying Neuropathology. Neurotherapeutics 2021; 18:728-752. [PMID: 34389969 PMCID: PMC8423978 DOI: 10.1007/s13311-021-01101-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia encompasses a group of clinical syndromes defined pathologically by degeneration of the frontal and temporal lobes. Historically, these syndromes have been challenging to diagnose, with an average of about three years between the time of symptom onset and the initial evaluation and diagnosis. Research in the field of neuroimaging has revealed numerous biomarkers of the various frontotemporal dementia syndromes, which has provided clinicians with a method of narrowing the differential diagnosis and improving diagnostic accuracy. As such, neuroimaging is considered a core investigative tool in the evaluation of neurodegenerative disorders. Furthermore, patterns of neurodegeneration correlate with the underlying neuropathological substrates of the frontotemporal dementia syndromes, which can aid clinicians in determining the underlying etiology and improve prognostication. This review explores the advancements in neuroimaging and discusses the phenotypic and pathologic features of behavioral variant frontotemporal dementia, semantic variant primary progressive aphasia, and nonfluent variant primary progressive aphasia, as seen on structural magnetic resonance imaging and positron emission tomography.
Collapse
Affiliation(s)
- Bradley T Peet
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nidhi Mundada
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| |
Collapse
|
39
|
Dev SI, Dickerson BC, Touroutoglou A. Neuroimaging in Frontotemporal Lobar Degeneration: Research and Clinical Utility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:93-112. [PMID: 33433871 PMCID: PMC8787866 DOI: 10.1007/978-3-030-51140-1_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Frontotemporal lobar dementia (FTLD) is a clinically and pathologically complex disease. Advances in neuroimaging techniques have provided a specialized set of tools to investigate underlying pathophysiology and identify clinical biomarkers that aid in diagnosis, prognostication, monitoring, and identification of appropriate endpoints in clinical trials. In this chapter, we review data discussing the utility of neuroimaging biomarkers in sporadic FTLD, with an emphasis on current and future clinical applications. Among those modalities readily utilized in clinical settings, T1-weighted structural magnetic resonance imaging (MRI) and 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) are best supported in differential diagnosis and as targets for clinical trial endpoints. However, a number of nonclinical neuroimaging modalities, including diffusion tensor imaging and resting-state functional connectivity MRI, show promise as biomarkers to predict progression and as clinical trial endpoints. Other neuroimaging modalities, including amyloid PET, Tau PET, and arterial spin labeling MRI, are also discussed, though more work is required to establish their utility in FTLD in clinical settings.
Collapse
Affiliation(s)
- Sheena I Dev
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, USA
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, USA.
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
40
|
Chu SA, Flagan TM, Staffaroni AM, Jiskoot LC, Deng J, Spina S, Zhang L, Sturm VE, Yokoyama JS, Seeley WW, Papma JM, Geschwind DH, Rosen HJ, Boeve BF, Boxer AL, Heuer HW, Forsberg LK, Brushaber DE, Grossman M, Coppola G, Dickerson BC, Bordelon YM, Faber K, Feldman HH, Fields JA, Fong JC, Foroud T, Gavrilova RH, Ghoshal N, Graff‐Radford NR, Hsiung GR, Huey ED, Irwin DJ, Kantarci K, Kaufer DI, Karydas AM, Knopman DS, Kornak J, Kramer JH, Kukull WA, Lapid MI, Litvan I, Mackenzie IR, Mendez MF, Miller BL, Onyike CU, Pantelyat AY, Rademakers R, Marisa Ramos E, Roberson ED, Carmela Tartaglia M, Tatton NA, Toga AW, Vetor A, Weintraub S, Wong B, Wszolek ZK, Van Swieten JC, Lee SE. Brain volumetric deficits in MAPT mutation carriers: a multisite study. Ann Clin Transl Neurol 2021; 8:95-110. [PMID: 33247623 PMCID: PMC7818091 DOI: 10.1002/acn3.51249] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/18/2020] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE MAPT mutations typically cause behavioral variant frontotemporal dementia with or without parkinsonism. Previous studies have shown that symptomatic MAPT mutation carriers have frontotemporal atrophy, yet studies have shown mixed results as to whether presymptomatic carriers have low gray matter volumes. To elucidate whether presymptomatic carriers have lower structural brain volumes within regions atrophied during the symptomatic phase, we studied a large cohort of MAPT mutation carriers using a voxelwise approach. METHODS We studied 22 symptomatic carriers (age 54.7 ± 9.1, 13 female) and 43 presymptomatic carriers (age 39.2 ± 10.4, 21 female). Symptomatic carriers' clinical syndromes included: behavioral variant frontotemporal dementia (18), an amnestic dementia syndrome (2), Parkinson's disease (1), and mild cognitive impairment (1). We performed voxel-based morphometry on T1 images and assessed brain volumetrics by clinical subgroup, age, and mutation subtype. RESULTS Symptomatic carriers showed gray matter atrophy in bilateral frontotemporal cortex, insula, and striatum, and white matter atrophy in bilateral corpus callosum and uncinate fasciculus. Approximately 20% of presymptomatic carriers had low gray matter volumes in bilateral hippocampus, amygdala, and lateral temporal cortex. Within these regions, low gray matter volumes emerged in a subset of presymptomatic carriers as early as their thirties. Low white matter volumes arose infrequently among presymptomatic carriers. INTERPRETATION A subset of presymptomatic MAPT mutation carriers showed low volumes in mesial temporal lobe, the region ubiquitously atrophied in all symptomatic carriers. With each decade of age, an increasing percentage of presymptomatic carriers showed low mesial temporal volume, suggestive of early neurodegeneration.
Collapse
Affiliation(s)
- Stephanie A. Chu
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Taru M. Flagan
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Adam M. Staffaroni
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lize C. Jiskoot
- Erasmus Medical CenterRotterdamNetherlands
- Dementia Research CenterUniversity College LondonLondonUK
| | - Jersey Deng
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Salvatore Spina
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Liwen Zhang
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Virginia E. Sturm
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Jennifer S. Yokoyama
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - William W. Seeley
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | | | - Howard J. Rosen
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Adam L. Boxer
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Hilary W. Heuer
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | | | - Murray Grossman
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | | | | | - Kelley Faber
- School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | | | - Jamie C. Fong
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Tatiana Foroud
- School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Nupur Ghoshal
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | | | - Edward D. Huey
- Departments of Psychiatry and NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - David J. Irwin
- University of California, Los AngelesLos AngelesCaliforniaUSA
| | - Kejal Kantarci
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Anna M. Karydas
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - John Kornak
- Department of Epidemiology and BiostatisticsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Joel H. Kramer
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Walter A. Kukull
- National Alzheimer's Coordinating CenterUniversity of WashingtonSeattleWashingtonUSA
| | | | - Irene Litvan
- University of California, San DiegoLa JollaCaliforniaUSA
| | | | - Mario F. Mendez
- University of California, Los AngelesLos AngelesCaliforniaUSA
| | - Bruce L. Miller
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | | | | | | | | | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative DiseasesUniversity of TorontoTorontoOntarioCanada
| | - Nadine A. Tatton
- The Association for Frontotemporal DegenerationRadnorPennsylvaniaUSA
| | - Arthur W. Toga
- USC Mark and Mary Stevens Neuroimaging and Informatics InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Ashley Vetor
- School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Bonnie Wong
- Massachusetts General HospitalBostonMassachusettsUSA
| | | | | | | | - Suzee E. Lee
- Memory and Aging CenterDepartment of Neurology, Weill Institute for NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
41
|
Soleimani-Meigooni DN, Iaccarino L, La Joie R, Baker S, Bourakova V, Boxer AL, Edwards L, Eser R, Gorno-Tempini ML, Jagust WJ, Janabi M, Kramer JH, Lesman-Segev OH, Mellinger T, Miller BL, Pham J, Rosen HJ, Spina S, Seeley WW, Strom A, Grinberg LT, Rabinovici GD. 18F-flortaucipir PET to autopsy comparisons in Alzheimer's disease and other neurodegenerative diseases. Brain 2020; 143:3477-3494. [PMID: 33141172 PMCID: PMC7719031 DOI: 10.1093/brain/awaa276] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022] Open
Abstract
Few studies have evaluated the relationship between in vivo18F-flortaucipir PET and post-mortem pathology. We sought to compare antemortem 18F-flortaucipir PET to neuropathology in a consecutive series of patients with a broad spectrum of neurodegenerative conditions. Twenty patients were included [mean age at PET 61 years (range 34-76); eight female; median PET-to-autopsy interval of 30 months (range 4-59 months)]. Eight patients had primary Alzheimer's disease pathology, nine had non-Alzheimer tauopathies (progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, and frontotemporal lobar degeneration with MAPT mutations), and three had non-tau frontotemporal lobar degeneration. Using an inferior cerebellar grey matter reference, 80-100-min 18F-flortaucipir PET standardized uptake value ratio (SUVR) images were created. Mean SUVRs were calculated for progressive supranuclear palsy, corticobasal degeneration, and neurofibrillary tangle Braak stage regions of interest, and these values were compared to SUVRs derived from young, non-autopsy, cognitively normal controls used as a standard for tau negativity. W-score maps were generated to highlight areas of increased tracer retention compared to cognitively normal controls, adjusting for age as a covariate. Autopsies were performed blinded to PET results. There was excellent correspondence between areas of 18F-flortaucipir retention, on both SUVR images and W-score maps, and neurofibrillary tangle distribution in patients with primary Alzheimer's disease neuropathology. Patients with non-Alzheimer tauopathies and non-tau frontotemporal lobar degeneration showed a range of tracer retention that was less than Alzheimer's disease, though higher than age-matched, cognitively normal controls. Overall, binding across both tau-positive and tau-negative non-Alzheimer disorders did not reliably correspond with post-mortem tau pathology. 18F-flortaucipir SUVRs in subcortical regions were higher in autopsy-confirmed progressive supranuclear palsy and corticobasal degeneration than in controls, but were similar to values measured in Alzheimer's disease and tau-negative neurodegenerative pathologies. Quantification of 18F-flortaucipir SUVR images at Braak stage regions of interest reliably detected advanced Alzheimer's (Braak VI) pathology. However, patients with earlier Braak stages (Braak I-IV) did not show elevated tracer uptake in these regions compared to young, tau-negative controls. In summary, PET-to-autopsy comparisons confirm that 18F-flortaucipir PET is a reliable biomarker of advanced Braak tau pathology in Alzheimer's disease. The tracer cannot reliably differentiate non-Alzheimer tauopathies and may not detect early Braak stages of neurofibrillary tangle pathology.
Collapse
Affiliation(s)
- David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Suzanne Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Viktoriya Bourakova
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rana Eser
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | | | - William J Jagust
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Mustafa Janabi
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Orit H Lesman-Segev
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Taylor Mellinger
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Julie Pham
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
42
|
Häkkinen S, Chu SA, Lee SE. Neuroimaging in genetic frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2020; 145:105063. [PMID: 32890771 DOI: 10.1016/j.nbd.2020.105063] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) have a strong clinical, genetic and pathological overlap. This review focuses on the current understanding of structural, functional and molecular neuroimaging signatures of genetic FTD and ALS. We overview quantitative neuroimaging studies on the most common genes associated with FTD (MAPT, GRN), ALS (SOD1), and both (C9orf72), and summarize visual observations of images reported in the rarer genes (CHMP2B, TARDBP, FUS, OPTN, VCP, UBQLN2, SQSTM1, TREM2, CHCHD10, TBK1).
Collapse
Affiliation(s)
- Suvi Häkkinen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie A Chu
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Suzee E Lee
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
43
|
Update on PET in neurodegenerative and neuroinflammatory disorders manifesting on a behavioural level: imaging for differential diagnosis. Curr Opin Neurol 2020; 32:548-556. [PMID: 31107281 DOI: 10.1097/wco.0000000000000706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW To give an update on recent findings concerning the use of PET for differential diagnosis in neurodegenerative and neuroinflammatory disorders manifesting on a behavioural level. RECENT FINDINGS Although accurate differential diagnosis of dementia can be achieved by imaging disease-specific patterns of cerebral glucose metabolism with [F]fluorodeoxyglucose ([F]FDG)-PET, the diagnostic impact of [F]FDG-PET in primary psychiatric disorders is limited. Amyloid-beta PET provides an incremental value beyond [F]FDG-PET in the differential diagnosis of dementia and was proposed as a biomarker defining the so-called Alzheimer continuum. Recently developed tau-specific tracers might also aid in the diagnostic process (biological definition of Alzheimer's disease together with amyloid-beta). Surpassing the diagnostic accuracy of other techniques, such as MRI, [F]FDG-PET has also gained widespread clinical use for diagnosis and follow-up of paraneoplastic and autoimmune disorders of the central nervous system (CNS) as an important differential diagnosis for rapid progressive dementia and subacute onset of psychiatric syndromes. SUMMARY Molecular neuroimaging with PET is an established method for the differential diagnosis of neurodegenerative and autoimmune CNS disorders manifesting on a behavioural level with significant therapeutic and prognostic impact. Future prospective studies are needed to define the value of tau imaging for diagnosis and prognosis in neurodegenerative disorders.
Collapse
|
44
|
Shen XN, Miao D, Li JQ, Tan CC, Cao XP, Tan L, Yu JT. MAPT rs242557 variant is associated with hippocampus tau uptake on 18F-AV-1451 PET in non-demented elders. Aging (Albany NY) 2020; 11:874-884. [PMID: 30708351 PMCID: PMC6382414 DOI: 10.18632/aging.101783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/15/2019] [Indexed: 11/29/2022]
Abstract
The microtubule-associated protein tau gene (MAPT) rs242557 variant is associated with multiple tauopathies and dementia. This study investigated whether it was correlated with brain tau-PET uptake in non-demented elders. Ninety non-demented elders were identified from the Alzheimer's Disease Neuroimaging Initiative cohort. We compared standardized uptake value ratios (SUVRs) of tau-PET tracer 18F-AV-1451 between rs242557 variant carriers and non-carriers in 25 regions of interest (ROIs). The minor allele A was associated with increased hippocampus 18F-AV-1451 uptake in non-demented elders (left: β = 0.111, Bonferroni corrected p = 0.035; right: β = 0.103, Bonferroni corrected p = 0.031). Aβ-positive participants (left: β = 0.206, Bonferroni corrected p = 0.029; right: β = 0.198, Bonferroni corrected p = 0.035) and APOE ε4 non-carriers (left: β = 0.140, Bonferroni corrected p = 0.006; right: β = 0.134, Bonferroni corrected p = 0.004) exhibited approximately the same findings in hippocampus. Considering no obvious associations in other regions, we confirmed the significant correlation of MAPT rs242557 risk variant with increased hippocampus tau deposition in non-demented elders. With higher magnitude signals in the hippocampus that is more likely to be uniquely affected in AD, the tau PET ligand 18F-AV-1451 seemed to possess a specific binding property for AD-like tau pathology.
Collapse
Affiliation(s)
- Xue-Ning Shen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Neurology, Fudan University, Shanghai, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Dan Miao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jie-Qiong Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Neurology, Fudan University, Shanghai, China.,Alzheimer's Disease Neuroimaging Initiative
| |
Collapse
|
45
|
Bevan-Jones WR, Cope TE, Jones PS, Kaalund SS, Passamonti L, Allinson K, Green O, Hong YT, Fryer TD, Arnold R, Coles JP, Aigbirhio FI, Larner AJ, Patterson K, O’Brien JT, Rowe JB. Neuroinflammation and protein aggregation co-localize across the frontotemporal dementia spectrum. Brain 2020; 143:1010-1026. [PMID: 32179883 PMCID: PMC7089669 DOI: 10.1093/brain/awaa033] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 12/04/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022] Open
Abstract
The clinical syndromes of frontotemporal dementia are clinically and neuropathologically heterogeneous, but processes such as neuroinflammation may be common across the disease spectrum. We investigated how neuroinflammation relates to the localization of tau and TDP-43 pathology, and to the heterogeneity of clinical disease. We used PET in vivo with (i) 11C-PK-11195, a marker of activated microglia and a proxy index of neuroinflammation; and (ii) 18F-AV-1451, a radioligand with increased binding to pathologically affected regions in tauopathies and TDP-43-related disease, and which is used as a surrogate marker of non-amyloid-β protein aggregation. We assessed 31 patients with frontotemporal dementia (10 with behavioural variant, 11 with the semantic variant and 10 with the non-fluent variant), 28 of whom underwent both 18F-AV-1451 and 11C-PK-11195 PET, and matched control subjects (14 for 18F-AV-1451 and 15 for 11C-PK-11195). We used a univariate region of interest analysis, a paired correlation analysis of the regional relationship between binding distributions of the two ligands, a principal component analysis of the spatial distributions of binding, and a multivariate analysis of the distribution of binding that explicitly controls for individual differences in ligand affinity for TDP-43 and different tau isoforms. We found significant group-wise differences in 11C-PK-11195 binding between each patient group and controls in frontotemporal regions, in both a regions-of-interest analysis and in the comparison of principal spatial components of binding. 18F-AV-1451 binding was increased in semantic variant primary progressive aphasia compared to controls in the temporal regions, and both semantic variant primary progressive aphasia and behavioural variant frontotemporal dementia differed from controls in the expression of principal spatial components of binding, across temporal and frontotemporal cortex, respectively. There was a strong positive correlation between 11C-PK-11195 and 18F-AV-1451 uptake in all disease groups, across widespread cortical regions. We confirmed this association with post-mortem quantification in 12 brains, demonstrating strong associations between the regional densities of microglia and neuropathology in FTLD-TDP (A), FTLD-TDP (C), and FTLD-Pick's. This was driven by amoeboid (activated) microglia, with no change in the density of ramified (sessile) microglia. The multivariate distribution of 11C-PK-11195 binding related better to clinical heterogeneity than did 18F-AV-1451: distinct spatial modes of neuroinflammation were associated with different frontotemporal dementia syndromes and supported accurate classification of participants. These in vivo findings indicate a close association between neuroinflammation and protein aggregation in frontotemporal dementia. The inflammatory component may be important in shaping the clinical and neuropathological patterns of the diverse clinical syndromes of frontotemporal dementia.
Collapse
Affiliation(s)
| | - Thomas E Cope
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sanne S Kaalund
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Luca Passamonti
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Consiglio Nazionale delle Ricerche (CNR), via Fratelli Cervi, Milano, Italy
| | - Kieren Allinson
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridge, UK
| | - Oliver Green
- Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Consiglio Nazionale delle Ricerche (CNR), via Fratelli Cervi, Milano, Italy
| | - Young T Hong
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Robert Arnold
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | | | | | | | - Karalyn Patterson
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B Rowe
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
46
|
Thijssen EH, La Joie R, Wolf A, Strom A, Wang P, Iaccarino L, Bourakova V, Cobigo Y, Heuer H, Spina S, VandeVrede L, Chai X, Proctor NK, Airey DC, Shcherbinin S, Duggan Evans C, Sims JR, Zetterberg H, Blennow K, Karydas AM, Teunissen CE, Kramer JH, Grinberg LT, Seeley WW, Rosen H, Boeve BF, Miller BL, Rabinovici GD, Dage JL, Rojas JC, Boxer AL. Diagnostic value of plasma phosphorylated tau181 in Alzheimer's disease and frontotemporal lobar degeneration. Nat Med 2020; 26:387-397. [PMID: 32123386 PMCID: PMC7101073 DOI: 10.1038/s41591-020-0762-2] [Citation(s) in RCA: 501] [Impact Index Per Article: 100.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/10/2020] [Indexed: 12/19/2022]
Abstract
With the potential development of new disease-modifying Alzheimer's disease (AD) therapies, simple, widely available screening tests are needed to identify which individuals, who are experiencing symptoms of cognitive or behavioral decline, should be further evaluated for initiation of treatment. A blood-based test for AD would be a less invasive and less expensive screening tool than the currently approved cerebrospinal fluid or amyloid β positron emission tomography (PET) diagnostic tests. We examined whether plasma tau phosphorylated at residue 181 (pTau181) could differentiate between clinically diagnosed or autopsy-confirmed AD and frontotemporal lobar degeneration. Plasma pTau181 concentrations were increased by 3.5-fold in AD compared to controls and differentiated AD from both clinically diagnosed (receiver operating characteristic area under the curve of 0.894) and autopsy-confirmed frontotemporal lobar degeneration (area under the curve of 0.878). Plasma pTau181 identified individuals who were amyloid β-PET-positive regardless of clinical diagnosis and correlated with cortical tau protein deposition measured by 18F-flortaucipir PET. Plasma pTau181 may be useful to screen for tau pathology associated with AD.
Collapse
Affiliation(s)
- Elisabeth H Thijssen
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Amy Wolf
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Ping Wang
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Viktoriya Bourakova
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Yann Cobigo
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Hilary Heuer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Lawren VandeVrede
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Xiyun Chai
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | | | | | - John R Sims
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Anna M Karydas
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Howie Rosen
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | | | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | | | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
47
|
Chen Q, Kantarci K. Imaging Biomarkers for Neurodegeneration in Presymptomatic Familial Frontotemporal Lobar Degeneration. Front Neurol 2020; 11:80. [PMID: 32184751 PMCID: PMC7058699 DOI: 10.3389/fneur.2020.00080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/22/2020] [Indexed: 02/05/2023] Open
Abstract
Frontotemporal lobar degeneration (FTLD) is a neurodegenerative disorder characterized by behavioral changes, language abnormality, as well as executive function deficits and motor impairment. In about 30-50% of FTLD patients, an autosomal dominant pattern of inheritance was found with major mutations in the MAPT, GRN, and the C9orf72 repeat expansion. These mutations could lead to neurodegenerative pathology years before clinical symptoms onset. With potential disease-modifying treatments that are under development, non-invasive biomarkers that help determine the early brain changes in presymptomatic FTLD patients will be critical for tracking disease progression and enrolling the right participants into the clinical trials at the right time during the disease course. In recent years, there is increasing evidence that a number of imaging biomarkers show the abnormalities during the presymptomatic stage. Imaging biomarkers of presymptomatic familial FTLD may provide insight into the underlying neurodegenerative process years before symptom onset. Structural magnetic resonance imaging (MRI) has demonstrated cortical degeneration with a mutation-specific neurodegeneration pattern years before onset of clinical symptoms in presymptomatic familial FTLD mutation carriers. In addition, diffusion tensor imaging (DTI) has shown the loss of white matter microstructural integrity in the presymptomatic stage of familial FTLD. Furthermore, proton magnetic resonance spectroscopy (1H MRS), which provides a non-invasive measurement of brain biochemistry, has identified early neurochemical abnormalities in presymptomatic MAPT mutation carriers. Positron emission tomography (PET) imaging with [18F]-fluorodeoxyglucose (FDG) has demonstrated the glucose hypometabolism in the presymptomatic stage of familial FTLD. Also, a novel PET ligand, 18F-AV-1451, has been used in this group to evaluate tau deposition in the brain. Promising imaging biomarkers for presymptomatic familial FTLD have been identified and assessed for specificity and sensitivity for accurate prediction of symptom onset and tracking disease progression during the presymptomatic stage when clinical measures are not useful. Furthermore, identifying imaging biomarkers for the presymptomatic stage is important for the design of disease-modifying trials. We review the recent progress in imaging biomarkers of the presymptomatic phase of familial FTLD and discuss the imaging techniques and analysis methods, with a focus on the potential implication of these imaging techniques and their utility in specific mutation types.
Collapse
Affiliation(s)
- Qin Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China.,Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
48
|
Convery RS, Jiao J, Clarke MTM, Moore KM, Koriath CAM, Woollacott IOC, Weston PSJ, Gunn R, Rabiner I, Cash DM, Rossor MN, Warren JD, Fox NC, Ourselin S, Bocchetta M, Rohrer JD. Longitudinal ( 18F)AV-1451 PET imaging in a patient with frontotemporal dementia due to a Q351R MAPT mutation. J Neurol Neurosurg Psychiatry 2020; 91:106-108. [PMID: 31439620 PMCID: PMC7611461 DOI: 10.1136/jnnp-2019-320904] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/10/2019] [Accepted: 07/11/2019] [Indexed: 11/04/2022]
Affiliation(s)
- Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Jieqing Jiao
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, London, UK
| | - Mica T M Clarke
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Katrina M Moore
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Carolin A M Koriath
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Ione O C Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Philip S J Weston
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Roger Gunn
- Imanova Centre for Imaging Sciences, London, UK
| | | | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Martin N Rossor
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Dementia Research Institute at UCL, London, UK
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
49
|
Papanikolopoulou K, Skoulakis EMC. Altered Proteostasis in Neurodegenerative Tauopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:177-194. [PMID: 32274757 DOI: 10.1007/978-3-030-38266-7_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Tauopathies are a heterogeneous group of neurodegenerative dementias involving perturbations in the levels, phosphorylation or mutations of the neuronal microtubule-binding protein Tau. Tauopathies are characterized by accumulation of hyperphosphorylated Tau leading to formation of a range of aggregates including macromolecular ensembles such as Paired Helical filaments and Neurofibrilary Tangles whose morphology characterizes and differentiates these disease states. Why nonphysiological Tau proteins elude the surveillance normal proteostatic mechanisms and eventually form these macromolecular assemblies is a central mostly unresolved question of cardinal importance for diagnoses and potential therapeutic interventions. We discuss the response of the Ubiquitin-Proteasome system, autophagy and the Endoplasmic Reticulum-Unfolded Protein response in Tauopathy models and patients, revealing interactions of components of these systems with Tau, but also of the effects of pathological Tau on these systems which eventually lead to Tau aggregation and accumulation. These interactions point to potential disease biomarkers and future potential therapeutic targets.
Collapse
Affiliation(s)
- Katerina Papanikolopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre "Alexander Fleming", Vari, Greece
| | - Efthimios M C Skoulakis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre "Alexander Fleming", Vari, Greece.
| |
Collapse
|
50
|
Llibre-Guerra JJ, Li Y, Schindler SE, Gordon BA, Fagan AM, Morris JC, Benzinger TLS, Hassenstab J, Wang G, Allegri R, Berman SB, Chhatwal J, Farlow MR, Holtzman DM, Jucker M, Levin J, Noble JM, Salloway S, Schofield P, Karch C, Fox NC, Xiong C, Bateman RJ, McDade E. Association of Longitudinal Changes in Cerebrospinal Fluid Total Tau and Phosphorylated Tau 181 and Brain Atrophy With Disease Progression in Patients With Alzheimer Disease. JAMA Netw Open 2019; 2:e1917126. [PMID: 31825500 PMCID: PMC6991202 DOI: 10.1001/jamanetworkopen.2019.17126] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPORTANCE The amyloid/tau/neurodegeneration (A/T/N) framework uses cerebrospinal fluid (CSF) levels of total tau (tTau) as a marker of neurodegeneration and CSF levels of phosphorylated tau 181 (pTau181) as a marker of tau tangles. However, it is unclear whether CSF levels of tTau and pTau181 have similar or different trajectories over the course of Alzheimer disease. OBJECTIVES To examine the rates of change in CSF levels of tTau and pTau181 across the Alzheimer disease course and how the rates of change are associated with brain atrophy as measured by magnetic resonance imaging. DESIGN, SETTING, AND PARTICIPANTS This cohort study was set in tertiary research clinics. Each participant was a member of a pedigree with a known mutation for dominantly inherited Alzheimer disease. Participants were divided into 3 groups on the basis of the presence of a mutation and their Clinical Dementia Rating score. Data analysis was performed in June 2019. MAIN OUTCOMES AND MEASURES Rates of change of CSF tTau and pTau181 levels and their association with the rate of change of brain volume. RESULTS Data from 465 participants (283 mutation carriers and 182 noncarriers) were analyzed. The mean (SD) age of the cohort was 37.8 (11.3) years, and 262 (56.3%) were women. The mean (SD) follow-up duration was 2.7 (1.5) years. Two or more longitudinal CSF and magnetic resonance imaging assessments were available for 160 and 247 participants, respectively. Sixty-five percent of mutation carriers (183) did not have symptoms at baseline (Clinical Dementia Rating score, 0). For mutation carriers, the annual rates of change for CSF tTau and pTau181 became significantly different from 0 approximately 10 years before the estimated year of onset (mean [SE] rates of change, 5.5 [2.8] for tTau [P = .05] and 0.7 [0.3] for pTau 181 [P = .04]) and 15 years before onset (mean [SE] rates of change, 5.4 [3.9] for tTau [P = .17] and 1.1 [0.5] for pTau181 [P = .03]), respectively. The rate of change of pTau181 was positive and increased at the early stages of the disease, showing a positive rate of change starting at 15 estimated years before onset until 5 estimated years before onset (mean [SE], 0.4 [0.3]), followed by a positive but decreasing rate of change at year 0 (mean [SE], 0.1 [0.3]) and then negative rates of change at 5 years (mean [SE], -0.3 [0.4]) and 10 years (mean [SE], -0.6 [0.6]) after symptom onset. In individuals without symptoms (Clinical Dementia Rating score, 0), the rates of change of CSF tTau and pTau181 were negatively associated with brain atrophy (high rates of change in CSF measures were associated with low rates of change in brain volume in asymptomatic stages). After symptom onset (Clinical Dementia Rating score, >0), an increased rate of brain atrophy was not associated with rates of change of levels of both CSF tTau and pTau181. CONCLUSIONS AND RELEVANCE These findings suggest that CSF tTau and pTau181 may have different associations with brain atrophy across the disease time course. These results have implications for understanding the dynamics of disease pathobiology and interpreting neuronal injury biomarker concentrations in response to Alzheimer disease progression and disease-modifying therapies.
Collapse
Affiliation(s)
| | - Yan Li
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | | | - Brian A. Gordon
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
| | - Anne M. Fagan
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - John C. Morris
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Hope Center for Neurological Disorders, St Louis, Missouri
- Knight Alzheimer’s Disease Research Center, St Louis, Missouri
| | | | - Jason Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Guoqiao Wang
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Tübingen, Germany
| | - Ricardo Allegri
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Sarah B. Berman
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | - David M. Holtzman
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Hope Center for Neurological Disorders, St Louis, Missouri
- Knight Alzheimer’s Disease Research Center, St Louis, Missouri
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Tübingen, Germany
- DZNE-German Center for Neurodegenerative Diseases, Tübingen, Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
- DZNE-German Center for Neurodegenerative Diseases, Munich, Munich, Germany
- SyNergy, Munich Cluster for Systems Neurology, Munich, Germany
| | - James M. Noble
- Taub Institute for Research on Alzheimer’s Disease, Aging Brain G.H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, New York
| | - Stephen Salloway
- Memory & Aging Program, Butler Hospital, Brown University, Providence, Rhode Island
| | - Peter Schofield
- Neuroscience Research Australia, Randwick, Sydney, New South Wales, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Celeste Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Nick C. Fox
- Dementia Research Centre, University College London, London, United Kingdom
| | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | - Randall J. Bateman
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| |
Collapse
|