1
|
Rabinovici GD, Selkoe DJ, Schindler SE, Aisen P, Apostolova LG, Atri A, Greenberg SM, Hendrix SB, Petersen RC, Weiner M, Salloway S, Cummings J. Donanemab: Appropriate use recommendations. J Prev Alzheimers Dis 2025; 12:100150. [PMID: 40155270 DOI: 10.1016/j.tjpad.2025.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Donanemab (Kisunla®), an IgG1 monoclonal antibody targeting N-terminal pyroglutamate-modified forms of amyloid-β, is approved in the United States for treatment of early symptomatic Alzheimer's disease (AD). Appropriate Use Recommendations (AUR) were developed to guide the implementation of donanemab in real-world practice, prioritizing safety considerations and opportunity for effectiveness. The AUR were developed by the AD and Related Disorders Therapeutic Workgroup by consensus, integrating available data and expert opinion. Appropriate candidates for donanemab treatment include persons with mild cognitive impairment or mild dementia due to AD (Clinical Stages 3-4, MMSE 20-30) who have biomarker confirmation of AD pathology by PET or CSF. Tau PET is not required for eligibility. Apolipoprotein E (APOE) genotyping should be performed prior to treatment to inform an individual's risk of developing Amyloid-Related Imaging Abnormalities (ARIA). Pre-treatment MRI should be obtained no more than 12 months prior to treatment. Patients with findings of >4 cerebral microbleeds, cortical superficial siderosis or a major vascular contribution to cognitive impairment should be excluded from treatment. The decision to initiate therapy should be grounded in a shared decision-making process that emphasizes the patient's values and goals of care. Donanemab is administered as a monthly intravenous infusion. Surveillance MRIs to evaluate for ARIA should be performed prior to the 2nd, 3rd, 4th and 7th infusions, prior to the 12th dose in higher risk individuals, and at any time ARIA is suspected clinically. Clinicians may consider discontinuing treatment if amyloid clearance is demonstrated by amyloid PET, typically obtained 12-18 months after initiating treatment.
Collapse
Affiliation(s)
- G D Rabinovici
- Memory & Aging Center, Departments of Neurology, Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - D J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - S E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - P Aisen
- Alzheimer's Treatment Research Institute, University of Southern California, San Diego, CA, USA
| | - L G Apostolova
- Departments of Neurology, Radiology, Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - A Atri
- Banner Sun Health Research Institute, Banner Health, Sun City, AZ, USA; Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - S M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - R C Petersen
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - M Weiner
- Departments of Radiology and Biomedical Imaging, Medicine, Psychiatry and Neurology, University of California San Francisco, San Francisco, CA, USA
| | - S Salloway
- Butler Hospital and Warren Alpert Medical School of Brown University, Providence RI, USA
| | - J Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
2
|
Ossenkoppele R, Salvadó G, Janelidze S, Pichet Binette A, Bali D, Karlsson L, Palmqvist S, Mattsson-Carlgren N, Stomrud E, Therriault J, Rahmouni N, Rosa-Neto P, Coomans EM, van de Giessen E, van der Flier WM, Teunissen CE, Jonaitis EM, Johnson SC, Villeneuve S, Benzinger TLS, Schindler SE, Bateman RJ, Doecke JD, Doré V, Feizpour A, Masters CL, Rowe C, Wiste HJ, Petersen RC, Jack CR, Hansson O. Plasma p-tau217 and tau-PET predict future cognitive decline among cognitively unimpaired individuals: implications for clinical trials. NATURE AGING 2025; 5:883-896. [PMID: 40155777 DOI: 10.1038/s43587-025-00835-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/12/2025] [Indexed: 04/01/2025]
Abstract
Plasma p-tau217 and tau positron emission tomography (PET) are strong prognostic biomarkers in Alzheimer's disease (AD), but their relative performance in predicting future cognitive decline among cognitively unimpaired (CU) individuals is unclear. In a head-to-head comparison study including nine cohorts and 1,474 individuals, we show that plasma p-tau217 and medial temporal lobe tau-PET signal display similar associations with cognitive decline on a global cognitive composite test (R2PET = 0.34 versus R2plasma = 0.33, Pdifference = 0.653) and with progression to mild cognitive impairment (hazard ratio (HR)PET = 1.61 (1.48-1.76) versus HRplasma = 1.57 (1.43-1.72), Pdifference = 0.322). Combined plasma and PET models were superior to the single-biomarker models (R2 = 0.35, P < 0.01). Sequential selection using plasma phosphorylated tau at threonine 217 (p-tau217) and then tau-PET reduced the number of participants required for a clinical trial by 94%, compared to a 76% reduction when using plasma p-tau217 alone. Thus, plasma p-tau217 and tau-PET showed similar performance for predicting future cognitive decline in CU individuals, and their sequential use enhances screening efficiency for preclinical AD trials.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Emma M Coomans
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Elsmarieke van de Giessen
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sylvia Villeneuve
- Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - 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, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - James D Doecke
- Australian eHealth Research Centre, Commonwealth Scientific and Industrial Research Organization, Melbourne, Victoria, Australia
| | - Vincent Doré
- Australian eHealth Research Centre, Commonwealth Scientific and Industrial Research Organization, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - Azadeh Feizpour
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Christopher Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
3
|
Bisi N, Pinzi L, Rastelli G. Selective imaging probes for differential detection of pathological tau polymorphs in tauopathies. Drug Discov Today 2025; 30:104352. [PMID: 40216294 DOI: 10.1016/j.drudis.2025.104352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/12/2025] [Accepted: 04/04/2025] [Indexed: 04/20/2025]
Abstract
Tauopathies, including Alzheimer's disease (AD), Pick's disease (PiD), progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD), are characterized by the misfolding and pathological aggregation of the tau protein, leading to neurodegeneration. Although the pathogenesis of these diseases is still a matter for debate, the formation of amyloid inclusions still represents the only histopathological hallmark available. Tau inclusions are not the same in terms of structure and morphology, and different tauopathies are characterized by different polymorphs. Remarkably, the selective detection of these polymorphs is crucial for differential diagnosis, disease monitoring and evaluation of the potential harmfulness of polymorphs, with a significant impact on drug discovery. This review discusses recent advances in the development of imaging probes designed for the selective detection of pathological tau forms associated with specific tauopathies. We explore the application of compounds that can target tau polymorphs characteristic of AD, PiD, PSP and CBD. In particular, we focus on discussing the probes' selectivity and sensitivity in distinguishing between the different tauopathy-associated polymorphs in preclinical settings. The progress and the weaknesses in this field are discussed, to guide the researchers in identifying accurate and potent probes for the selective diagnosis of these different neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicolò Bisi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy.
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
| |
Collapse
|
4
|
Salloway S, Pain A, Lee E, Papka M, Ferguson MB, Wang H, Hu H, Lu M, Oru E, Ardayfio PA, Hoban DB, Collins EC, Brooks DA, Sims JR. TRAILBLAZER-ALZ 4: A phase 3 trial comparing donanemab with aducanumab on amyloid plaque clearance in early, symptomatic Alzheimer's disease. Alzheimers Dement 2025; 21:e70293. [PMID: 40390253 PMCID: PMC12089073 DOI: 10.1002/alz.70293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/21/2025]
Abstract
INTRODUCTION The phase 3, open-label TRAILBLAZER-ALZ 4 study compared the effect of donanemab versus aducanumab on amyloid plaque (AP) clearance in participants with early symptomatic Alzheimer's disease. METHODS Participants (n = 148) were randomized 1:1 to receive intravenous donanemab (700 mg every 4 weeks for three doses, then 1400 mg every 4 weeks thereafter) or aducanumab (per label). AP was measured with florbetapir F 18 positron emission tomography. AP clearance was defined as < 24.1 Centiloids. RESULTS At 6, 12, and 18 months, AP clearance was achieved in 37.9%, 70.0%, and 76.8%, respectively, of donanemab-treated participants versus 1.6%, 24.6%, and 43.1% of aducanumab-treated participants (P < 0.001). Median time to clearance was 359 versus 568 days for donanemab- versus aducanumab-treated participants (P < 0.001). Amyloid-related imaging abnormality (ARIA)-edema/effusion occurred in 23.9% and 34.8% of donanemab- and aducanumab-treated participants, respectively. DISCUSSION Donanemab treatment resulted in earlier and greater AP clearance compared to aducanumab. ARIA frequencies were consistent with prior studies. CLINICAL TRIAL REGISTRATION No: NCT05108922, TRAILBLAZER-ALZ 4 HIGHLIGHTS: Here we report the first direct comparator study of two amyloid-targeting therapies. This was the first investigation of donanemab on biomarker efficacy regardless of tau levels. Donanemab demonstrated superiority over aducanumab in amyloid plaque (AP) clearance. The depth and speed of AP removal did not affect amyloid-related imaging abnormality risk or incidence.
Collapse
Affiliation(s)
- Stephen Salloway
- Department of Neurology and Department of PsychiatryAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Butler HospitalProvidenceRhode IslandUSA
| | | | - Elly Lee
- Irvine Clinical ResearchIrvineCaliforniaUSA
| | - Michelle Papka
- The Cognitive and Research Center of New Jersey LLCSpringfieldNew JerseyUSA
| | | | - Hong Wang
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Haoyan Hu
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Ming Lu
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Ena Oru
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | | | | | | | | | |
Collapse
|
5
|
Dresse MT, Ferreira PCL, Prasadan A, Diaz JL, Zeng X, Bellaver B, Povala G, Villemagne VL, Kamboh MI, Cohen AD, Pascoal TA, Ganguli M, Snitz BE, Shaaban CE, Karikari TK. Plasma biomarkers identify brain ATN abnormalities in a dementia-free population-based cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.27.25326360. [PMID: 40343022 PMCID: PMC12060967 DOI: 10.1101/2025.04.27.25326360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Using the ATN framework, we evaluated the potential of plasma biomarkers to identify abnormal brain amyloid-beta (Aβ) positron emission tomography (PET), tau-PET and neurodegeneration in a socioeconomically disadvantaged population-based cohort. METHODS Community-dwelling dementia-free (n=113, including 102 (91%) cognitively normal) participants underwent ATN neuroimaging and plasma biomarker assessments. RESULTS Plasma Aβ42/Aβ40, p-tau181, and p-tau217 showed significant associations with Aβ-PET status (adjusted odds ratio [AOR] of 1.74*10-24, 1.47, and 3.43*103 respectively [p-values<0.05]), with p-tau217 demonstrating the highest classification accuracy for Aβ-PET status (AUC=0.94). Plasma p-tau181 and p-tau217 showed significant associations with tau-PET status (AOR: 1.50 and 22.24, respectively, p-values<0.05), with comparable classification accuracies for tau-PET status (AUC=0.74 and 0.70, respectively). Only plasma NfL showed significant association with neurodegeneration based on cortical thickness (AOR=1.09, p-value<0.05). CONCLUSION Our findings highlight potential of plasma p-tau217 as a biomarker for brain Aβ and tau pathophysiology, p-tau181 for tau abnormalities, and NfL for neurodegeneration in the community.
Collapse
Affiliation(s)
- Menayit Tamrat Dresse
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Pamela C. L. Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Akshay Prasadan
- Department of Statistics & Data Science, Dietrich College of Humanities and Social Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Jihui L. Diaz
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Ilyas Kamboh
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ann D. Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mary Ganguli
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Beth E. Snitz
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - C. Elizabeth Shaaban
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
- Alzheimer’s Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Nasrallah IM, Kuo PH, Nordberg A, Bohnen NI, Ponisio MR. The Impact of Amyloid and Tau PET on Alzheimer Disease Diagnostics: AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2025. [PMID: 40237426 DOI: 10.2214/ajr.24.32325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Amyloid and tau PET have contributed significantly to understanding the biology of Alzheimer disease (AD), aided development of biomarker-driven AD diagnostic criteria, and facilitated approval of the first disease-modifying drugs for AD. As opportunities to use amyloid and tau PET in the clinic have expanded, several factors will impact their application and real-world impact in patients with AD. First, quantification of amyloid and tau PET interpretations, supported by appropriate visual confirmation, will be needed for monitoring therapy response. Also, amyloid and tau PET will need to be balanced with emerging biofluid assays from CSF and blood. Blood-based biomarkers, although still requiring validation, have particular potential to complement PET utilization; nonetheless, the topographic information uniquely provided by tau PET will remain important in clinical practice. Additionally, the proper use of amyloid and tau PET for clinical management will require an understanding and consideration of mixed pathology, as is usually present in AD, along with continued advances in imaging technology to better address copathology. Further research and investment in this evolving field will improve diagnostic accuracy and therapeutic approaches, ultimately benefiting outcomes in patients with AD.
Collapse
Affiliation(s)
- Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Phillip H Kuo
- Division of Nuclear Medicine, City of Hope® Cancer Center Duarte, Duarte, CA, USA
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institute, Stockholm, Sweden
| | - Nicolaas I Bohnen
- Departments of Radiology and Neurology, University of Michigan, and Neurology service and Geriatric Research, Education, and Clinical Center, Ann Arbor VA Medical Center, Ann Arbor, MI, USA
| | - Maria R Ponisio
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
7
|
Katsumi Y, Howe IA, Eckbo R, Wong B, Quimby M, Hochberg D, McGinnis SM, Putcha D, Wolk DA, Touroutoglou A, Dickerson BC. Default mode network tau predicts future clinical decline in atypical early Alzheimer's disease. Brain 2025; 148:1329-1344. [PMID: 39412999 PMCID: PMC11969453 DOI: 10.1093/brain/awae327] [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: 04/09/2024] [Revised: 08/31/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Identifying individuals with early-stage Alzheimer's disease (AD) at greater risk of steeper clinical decline would enable better-informed medical, support and life planning decisions. Despite accumulating evidence on the clinical prognostic value of tau PET in typical late-onset amnestic AD, its utility in predicting clinical decline in individuals with atypical forms of AD remains unclear. Across heterogeneous clinical phenotypes, patients with atypical AD consistently exhibit abnormal tau accumulation in the posterior nodes of the default mode network of the cerebral cortex. This evidence suggests that tau burden in this functional network could be a common imaging biomarker for prognostication across the syndromic spectrum of AD. Here, we examined the relationship between baseline tau PET signal and the rate of subsequent clinical decline in a sample of 48 A+/T+/N+ patients with mild cognitive impairment or mild dementia due to AD with atypical clinical phenotypes: Posterior Cortical Atrophy (n = 16); logopenic variant Primary Progressive Aphasia (n = 15); and amnestic syndrome with multi-domain impairment and young age of onset < 65 years (n = 17). All patients underwent MRI, tau PET and amyloid PET scans at baseline. Each patient's longitudinal clinical decline was assessed by calculating the annualized change in the Clinical Dementia Rating Sum-of-Boxes (CDR-SB) scores from baseline to follow-up (mean time interval = 14.55 ± 3.97 months). Atypical early AD patients showed an increase in CDR-SB by 1.18 ± 1.25 points per year: t(47) = 6.56, P < 0.001, Cohen's d = 0.95. Across clinical phenotypes, baseline tau in the default mode network was the strongest predictor of clinical decline (R2 = 0.30), outperforming a simpler model with baseline clinical impairment and demographic variables (R2 = 0.10), tau in other functional networks (R2 = 0.11-0.26) and the magnitude of cortical atrophy (R2 = 0.20) and amyloid burden (R2 = 0.09) in the default mode network. Overall, these findings point to the contribution of default mode network tau to predicting the magnitude of clinical decline in atypical early AD patients 1 year later. This simple measure could aid the development of a personalized prognostic, monitoring and treatment plan, which would help clinicians not only predict the natural evolution of the disease but also estimate the effect of disease-modifying therapies on slowing subsequent clinical decline given the patient's tau burden while still early in the disease course.
Collapse
Affiliation(s)
- Yuta Katsumi
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Alzheimer’s Disease Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Inola A Howe
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ryan Eckbo
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Bonnie Wong
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Megan Quimby
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Daisy Hochberg
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Scott M McGinnis
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Center for Brain/Mind Medicine, Department of Neurology, Brigham & Women’s Hospital, Boston, MA 02115, USA
| | - Deepti Putcha
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Center for Brain/Mind Medicine, Department of Neurology, Brigham & Women’s Hospital, Boston, MA 02115, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra Touroutoglou
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Alzheimer’s Disease Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Alzheimer’s Disease Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
8
|
Aiello M, Marizzoni M, Borrelli P, Cavaliere C, Ribaldi F, Garibotto V, Scheffler M, Jelescu IO, Jovicich J, Catani M, Salvatore M, Frisoni GB, Pievani M. Microstructural assessment of the locus coeruleus-entorhinal cortex pathway and association with ATN markers in cognitive impairment. Alzheimers Dement 2025; 21:e70126. [PMID: 40289861 PMCID: PMC12035542 DOI: 10.1002/alz.70126] [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: 10/16/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 04/30/2025]
Abstract
INTRODUCTION Whether Alzheimer's disease pathology involves white matter pathways connecting the locus coeruleus (LC) to the entorhinal cortex (EC) is unclear. In this cross-sectional observational study, we investigated the microstructural integrity of the LC-EC pathway in relation to amyloid, tau, and neurodegeneration (ATN) biomarkers along the cognitive spectrum from normal cognition to dementia. METHODS One hundred twenty-four participants underwent clinical assessment, diffusion-weighted imaging, structural magnetic resonance imaging (N), amyloid (A), and tau (T) positron emission tomography. Diffusivity indices were assessed in the LC-EC tract using a probabilistic atlas, and linear models were used to assess associations with ATN markers and cognition. RESULTS Differences in LC-EC microstructural parameters were observed in participants with Braak stage > I versus Braak 0 (p < 0.020), N+ versus N- (p < 0.001), and cognitively impaired versus unimpaired (p < 0.019). LC-EC mean diffusivity was associated with Mini-Mental State Examination score even after accounting for ATN markers (p = 0.015). DISCUSSION Our results suggest that LC-EC diffusivity provides complementary information over ATN biomarkers in explaining cognitive impairment. HIGHLIGHTS Locus coeruleus-entorhinal cortex (LC-EC) tract microstructure is associated with tau and especially neurodegeneration markers. LC-EC tract microstructure is more sensitive to tau pathology and neurodegeneration than tracts commonly affected in Alzheimer's disease. LC-EC diffusivity measures provide complementary information over amyloid, tau, and neurodegeneration (ATN) biomarkers.
Collapse
Affiliation(s)
| | - Moira Marizzoni
- Biological Psychiatry UnitIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | | | | | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE)University of GenevaGenevaSwitzerland
- Geneva Memory CenterDepartment of Rehabilitation and GeriatricsGeneva University HospitalsGenevaSwitzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab)Geneva University Neurocenter and Faculty of Medicine, University of GenevaGenevaSwitzerland
- Division of Nuclear Medicine and Molecular ImagingGeneva University HospitalsGenevaSwitzerland
- CIBM Center for Biomedical ImagingGenevaSwitzerland
| | - Max Scheffler
- Division of RadiologyGeneva University HospitalsGenevaSwitzerland
| | - Ileana O. Jelescu
- Lausanne University Hospital (CHUV) and University of Lausanne (UNIL)LausanneSwitzerland
| | - Jorge Jovicich
- Center for Mind/Brain SciencesUniversity of TrentoMattarelloItaly
| | | | | | - Giovanni B. Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE)University of GenevaGenevaSwitzerland
- Geneva Memory CenterDepartment of Rehabilitation and GeriatricsGeneva University HospitalsGenevaSwitzerland
| | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and EpidemiologyIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| |
Collapse
|
9
|
Rikken RM, Coomans EM, de Koning LA, Visser D, Neutelings E, den Braber A, Collij LE, Golla SSV, for the Alzheimer's Disease Neuroimaging Initiative, Barkhof F, Visser PJ, Scheltens P, van der Flier WM, Boellaard R, Ossenkoppele R, Vijverberg EGB, van de Giessen E, for ADNI. Characterizing visual read tau-PET-negative participants with Alzheimer's disease dementia. Alzheimers Dement 2025; 21:e14423. [PMID: 40219781 PMCID: PMC11992537 DOI: 10.1002/alz.14423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/09/2024] [Accepted: 11/01/2024] [Indexed: 04/14/2025]
Abstract
INTRODUCTION A subset of amyloid beta (Aβ)-positive Alzheimer's disease (AD) patients is tau-positron emission tomography (PET) negative. We aimed to characterize this subgroup using [18F]flortaucipir PET visual read (VR), as this is important for prognosis and selection for therapies. METHODS Aβ-positive VR tau-PET-negative AD dementia patients (AD A+T-) were compared to tau-PET-positive AD patients (AD A+T+) and control groups (CU A-T-; CU A+T-) included from the Amsterdam-based cohort and Alzheimer's Disease Neuroimaging Initiative (ADNI). We compared [18F]flortaucipir binding in an early- and late-stage tau ROI, atrophy, cognition, and co-pathologies. RESULTS AD A+T- were older, showed less hippocampal atrophy and slower cognitive decline compared to AD A+T+. In ADNI, AD A+T- showed higher early-stage tau binding compared to both control groups and more late-stage tau compared to CU A-T-, but no tau accumulation over time. DISCUSSION VR tau-PET-negative AD patients show neurodegenerative and cognitive processes consistent with the AD trajectory, but milder progression compared to tau-PET-positive AD patients. HIGHLIGHTS We used the novel Food and Drug Administration (FDA)-approved VR method for defining tau-PET positivity. AD A+T- patients were older and showed less atrophy and cognitive decline than AD A+T+. We did not find convincing evidence of tau accumulation in AD A+T- or copathologies. The group of AD A+T- patients is likely very heterogeneous.
Collapse
Affiliation(s)
- Roos M. Rikken
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Emma M. Coomans
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Lotte A. de Koning
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Denise Visser
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Eline Neutelings
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Biological PsychiatryVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Lyduine E. Collij
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Clinical Memory Research Unit, Clinical Sciences Malmö, Faculty of MedicineLund University, Skånes Universitetssjukhus, VE MinnessjukdomarMalmöSweden
| | - Sandeep S. V. Golla
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | | | - Frederik Barkhof
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image ComputingUniversity College LondonLondonUK
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Alzheimer Center Limburg, School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of NeurogeriatricsKarolinska InstitutetSolnaSweden
| | - Philip Scheltens
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Epidemiology & Data ScienceVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Ronald Boellaard
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdam UMC location VUmcAmsterdamThe Netherlands
- Clinical Memory Research Unit, Clinical Sciences Malmö, Faculty of MedicineLund University, Skånes Universitetssjukhus, VE MinnessjukdomarMalmöSweden
| | - Everard G. B. Vijverberg
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdam UMC location VUmcAmsterdamThe Netherlands
| | - Elsmarieke van de Giessen
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam UMC location VUmcAmsterdamThe Netherlands
| | | |
Collapse
|
10
|
Cai W, Neitzel J, Glodzik L, Blacker D, Ma Y. Effect Modifiers of the Association of Blood Pressure With Brain Amyloid and Tau Pathology. Neurology 2025; 104:e213441. [PMID: 40014836 PMCID: PMC11874733 DOI: 10.1212/wnl.0000000000213441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/15/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Hypertension is an important modifiable risk factor of Alzheimer disease (AD), but previous studies reported heterogeneous associations of late-life blood pressure (BP) with brain amyloid and tau pathologies. We investigated how the associations of BP with brain amyloid and tau vary by APOE ε4 carriership, age, cerebrovascular burden, and cognitive status. METHODS We performed analyses among participants with postmortem neuropathology measurements (2005-June 2022) from the National Alzheimer's Coordinating Center. The average systolic BP (SBP) of the first 3 annual visits was the primary exposure, and baseline hypertension status was the secondary exposure. Brain AD pathologies were assessed using Thal and Braak staging. Potential modifiers included APOE ε4 carriership, age, stroke history, and cognitive status. Multinomial logistic regressions with interaction terms were used to test effect modification, adjusting for age, sex, APOE ε4 carriership, education, antihypertensive medication use, and years to death. RESULTS Among 2,094 participants (baseline age: 75 ± 9.5 years; 51.4% women), the association of higher SBP with higher amyloid and tau burdens varied by stroke history and cognitive status while the effect modification by age or APOE ε4 carriership was less consistent. More pronounced associations of SBP with higher amyloid and tau burdens were observed in those with dementia (vs without dementia) and those with a history of stroke (vs without stroke) (All p interaction<0.05). The odds ratios (ORs) per 10-mm Hg increase in SBP in the stroke vs nonstroke subgroup were 1.58 (95% CI 1.04-2.41) vs 1.14 (1.03-1.27) for amyloid and 1.54 (1.00-2.36) vs 1.04 (0.96-1.12) for tau. When comparing dementia with cognitively normal subgroups, ORs were 1.39 (1.17-1.64) vs 1.12 (0.96-1.31) for amyloid and 1.24 (1.08-1.42) vs 0.98 (0.85-1.14) for tau. Similar findings were observed for baseline hypertension status. DISCUSSION Preexisting cerebrovascular burden and cognitive status might interact with elevated SBP in their association with higher brain amyloid and tau, which could help identify high-risk subgroups for BP management and AD prevention. These heterogeneous association patterns need to be confirmed in longitudinal studies with in vivo AD pathology assessments.
Collapse
Affiliation(s)
- Wenjie Cai
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Julia Neitzel
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Lidia Glodzik
- Department of Radiology, Weill Cornell Medicine, Brain Health Imaging Institute, New York, NY; and
| | - Deborah Blacker
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Yuan Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
11
|
Nie P, Wu Y, Robinson J, Mekala S, Lee VMY, Li YM. In Situ Labeling of Pathogenic Tau Using Photo-Affinity Chemical Probes. ACS Chem Biol 2025; 20:581-591. [PMID: 40079621 DOI: 10.1021/acschembio.5c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Tau aggregation plays a crucial role in the development of Alzheimer's disease (AD). Developing specific techniques that can isolate pathogenic tau from brain tissue is important for understanding tauopathies and advancing targeted therapies. Here, we develop photoaffinity small molecular probes and a novel method for in situ tissue labeling and investigate their activity in interacting with tau in cells and AD patient brains. Based on the reported chemical structures of tau PET tracers, we designed and synthesized two tau-specific probes, namely, Tau-2 and Tau-4. After validation in cell, mouse model, and patient brain samples, our photolabeling results suggested that Tau-2 effectively labels soluble tau in cell and mouse models, while Tau-4 selectively binds high-molecular-weight tau aggregates in late-stage AD patient brain tissues. Proteomic analysis verified the specific isolation of pathogenic tau from AD brain samples. Collectively, these findings underscore the potential of our photoaffinity probes as powerful tools for investigating tau proteins and neurofibrillary tangles in neurodegenerative diseases.
Collapse
Affiliation(s)
- Pengju Nie
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - You Wu
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - John Robinson
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Shekar Mekala
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - Virginia M Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| |
Collapse
|
12
|
Thal DR, Poesen K, Vandenberghe R, De Meyer S. Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers. Mol Neurodegener 2025; 20:33. [PMID: 40087672 PMCID: PMC11907863 DOI: 10.1186/s13024-025-00819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.
Collapse
Affiliation(s)
- Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.
| | - Koen Poesen
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Steffi De Meyer
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Tunali I, Wang J, Arora AK, Kim MJ, Shcherbinin S, Pontecorvo M, Iaccarino L. Development and Validation of a 18F-Flortaucipir PET Visual Stratification Method. J Nucl Med 2025; 66:jnumed.124.268700. [PMID: 40081955 PMCID: PMC11960607 DOI: 10.2967/jnumed.124.268700] [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: 08/28/2024] [Accepted: 01/14/2025] [Indexed: 03/16/2025] Open
Abstract
Tau PET quantitation methods have been used in research settings and clinical trials to measure tau burden for diagnostic, staging, and prognostic purposes. However, these methods require specialized software, skilled analysts, and advanced image processing. We developed a novel 18F-flortaucipir PET (FTP, or Tauvid) visual read method enabling stratification of patients with Alzheimer disease (AD) according to the tau level without the need for quantitation. An independent reader study (I7E-AV-A26) was conducted to test this method against a quantitation-based high-tau standard of truth. Methods: A total of 140 baseline or screening FTP scans were randomly selected from the TRAILBLAZER-ALZ 2 phase 3 trial (NCT04437511). Five qualified imaging physicians were trained for the FTP visual stratification method, using previously identified thresholds and cortical regions of interest thought to optimally stratify high-tau and non-high-tau scans. Positive and negative percent agreement (PPA and NPA, respectively) between visual stratifications and quantitation-based high tau (AD-signature SUV ratio > 1.46) were calculated. Predefined success criteria were met if the lower bounds of a 2-sided 95% CI for PPA and NPA were 50% or greater for at least 3 of the 5 readers. Inter- and intrareader reliability were assessed using Fleiss κ (n = 140) and Cohen κ (n = 20 test-retest scans) metrics. Results: The median PPA and NPA were 83.4% and 88.9%, respectively, with lower bounds of 2-sided 95% CIs being 50% or greater for all readers. The Fleiss κ-point estimate was 0.8882 (95% CI, 0.8356-0.9409) and the Cohen κ-point estimate was 0.9599 (95% CI, 0.9049-1.000) for all readers, indicating almost perfect inter- and intrareader agreement. Study I7E-AV-A26 successfully validated the feasibility of the FTP visual stratification method, possibly supporting AD staging and prognosis with high inter- and intrareader agreements, confirming the reliability of the method. Conclusion: Future investigations may include expanding the validation dataset, including real-world clinical data from diverse populations, using autopsy confirmation, exploring alternative regions and thresholds for other tau PET stratifications, and assessing differences in treatment response among visually stratified participants enrolled in disease-modifying therapy trials.
Collapse
Affiliation(s)
- Ilke Tunali
- Eli Lilly and Company, Indianapolis, Indiana; and
| | - Jian Wang
- Eli Lilly and Company, Indianapolis, Indiana; and
| | | | - Min Jung Kim
- Eli Lilly and Company, Indianapolis, Indiana; and
| | | | | | - Leonardo Iaccarino
- Eli Lilly and Company, Indianapolis, Indiana; and
- Eli Lilly Italia S.p.A, Sesto Fiorentino, Italy
| |
Collapse
|
14
|
De Sousa RAL, Mendes BF. T-regulatory cells and extracellular vesicles in Alzheimer's disease: New therapeutic concepts and hypotheses. Brain Res 2025; 1850:149393. [PMID: 39672489 DOI: 10.1016/j.brainres.2024.149393] [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: 05/13/2024] [Revised: 10/27/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Cell-based treatment has experienced exponential expansion in recent years in terms of clinical application and market share among pharmaceutical companies. When malignant cells in a healthy individual produce antigenic peptides derived from mutant or improperly synthesized proteins, the immune system attacks and kills the transforming cells. This process is carried out continuously by immune cells scanning the body for altered cells that could cause some harm. T-regulatory cells (Tregs), which preserve immunological tolerance and can exert neuroprotective benefits in numerous disorders, including animal models of Alzheimer's disease (AD), have demonstrated considerable therapeutic potential. Evidence also suggests that not only Tregs, but extracellular vesicles (EVs) are involved in a wide range of diseases, such as cellular homoeostasis, infection propagation, cancer development and heart disease, and have become a promisor cell-based therapeutic field too. Nevertheless, despite significant recent clinical and commercial breakthroughs, cell-based medicines still confront numerous challenges that hinder their general translation and commercialization. These challenges include, but are not limited to, choosing the best cell source, and creating a product that is safe, adequately viable, and fits the needs of individual patients and diseases. Here, we summarize what we know about Tregs and EVs and their potential therapeutic usage in AD.
Collapse
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Physical Education Department, Federal University of the Valleys of Jequitinhonha and Mucuri (UFVJM), Diamantina, MG, Brazil.
| | - Bruno Ferreira Mendes
- Physical Education Department, Federal University of the Valleys of Jequitinhonha and Mucuri (UFVJM), Diamantina, MG, Brazil; Physical Education Department, UNIPTAN, São João Del Rey, MG, Brazil
| |
Collapse
|
15
|
Völter F, Eckenweber S, Scheifele M, Eckenweber F, Hirsch F, Franzmeier N, Kreuzer A, Griessl M, Steward A, Janowitz D, Palleis C, Bernhardt A, Vöglein J, Stockbauer A, Rauchmann BS, Schöberl F, Wlasich E, Buerger K, Wagemann O, Perneczky R, Weidinger E, Höglinger G, Levin J, Brendel M, Schönecker S. Correlation of early-phase β-amyloid positron-emission-tomography and neuropsychological testing in patients with Alzheimer's disease. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07175-5. [PMID: 40019578 DOI: 10.1007/s00259-025-07175-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Clinical staging in individuals with Alzheimer's disease (AD) typically relies on neuropsychological testing. Recognizing the imperative for an objective measure of clinical AD staging, regional perfusion in early-phase β-amyloid-PET may aid as a cost-efficient index for the assessment of neurodegeneration severity in patients with Alzheimer's disease. METHODS Regional perfusion deficits in early-phase β-amyloid-PET as well as neuropsychological testing (max. 90 days delay) were evaluated in 82 patients with biologically defined AD according to the ATN classification. In reference to the Braak staging system patients were classified into the groups stage0, stageI-II+, stageI-IV+, stageI-VI+, and stageatypical+ according to regional perfusion deficits in regions of interest (ROIs) published by the Alzheimer's Disease Neuroimaging Initiative. Multiple regression analysis controlling for age, gender, and education was used to evaluate the association of regional z-scores on perfusion-phase PET with clinical scores for all patients and with annual decline of cognitive performance in 23 patients with follow-up data. RESULTS Patients classified as stage0 and stageI-II+ demonstrated significantly superior neuropsychological performance compared to those classified as stageI-IV+ and stageI-VI+. Lower cognitive performance was associated with decreased perfusion in early-phase β-amyloid-PET globally and regionally, with the most pronounced association identified in the left temporal lobe. Mean z-scores on early-phase PET in temporal and parietal regions offered a robust prediction of future annual decline in MMSE and sum scores of the CERAD-Plus (Consortium to Establish a Registry for Alzheimer's Disease) test battery. CONCLUSION Regional and global perfusion deficits in early-phase β-amyloid-PET can serve as an objective index of neurodegeneration severity and may act as prognostic markers of future cognitive decline in AD.
Collapse
Affiliation(s)
- Friederike Völter
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany.
| | - Sebastian Eckenweber
- 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
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Fabian Hirsch
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Annika Kreuzer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Maria Griessl
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Carla Palleis
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Alexander Bernhardt
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jonathan Vöglein
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Anna Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Neuroradiology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Florian Schöberl
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elisabeth Wlasich
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Olivia Wagemann
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Endy Weidinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Günter Höglinger
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Johannes Levin
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Sonja Schönecker
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
16
|
Alosco ML, Mejía Pérez J, Culhane JE, Shankar R, Nowinski CJ, Bureau S, Mundada N, Smith K, Amuiri A, Asken B, Groh JR, Miner A, Pettway E, Mosaheb S, Tripodis Y, Windon C, Mercier G, Stern RA, Grinberg LT, Soleimani-Meigooni DN, Christian BT, Betthauser TJ, Stein TD, McKee AC, Mathis CA, Abrahamson EE, Ikonomovic MD, Johnson SC, Mez J, La Joie R, Schonhaut D, Rabinovici GD. 18F-MK-6240 tau PET in patients at-risk for chronic traumatic encephalopathy. Mol Neurodegener 2025; 20:23. [PMID: 39994806 PMCID: PMC11852567 DOI: 10.1186/s13024-025-00808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Molecular biomarkers of chronic traumatic encephalopathy (CTE) are lacking. We evaluated 18F-MK-6240 tau PET as a biomarker for CTE. Two studies were done: (1) 3H-MK-6240 autoradiography and an in-vitro brain homogenate binding studies on postmortem CTE tissue, (2) an in-vivo 18F-MK-6240 tau PET study in former American football players. METHODS Autoradiography and in-vitro binding studies were done using 3H-MK-6240 on frozen temporal and frontal cortex tissue from six autopsy cases with stage III CTE compared to Alzheimer's disease. Thirty male former National Football League (NFL) players with cognitive concerns (mean age = 58.9, SD = 7.8) completed tau (18F-MK-6240) and Aβ (18F-Florbetapir) PET. Controls included 39 Aβ-PET negative, cognitively normal males (mean age = 65.7, SD = 6.3). 18F-MK-6240 SUVr images were created using 70-90 min post-injection data with inferior cerebellar gray matter as the reference. We compared SUVr between players and controls using voxelwise and region-of-interest approaches. Correlations between 18F-MK-6240 SUVr and cognitive scores were tested. RESULTS All six CTE stage III cases had Braak NFT stage III but no neuritic plaques. Two had Thal Phase 1 for Aβ; one showed a laminar pattern of 3H-MK-6240 autoradiography binding in the superior temporal cortex and less so in the dorsolateral frontal cortex, corresponding to tau-immunoreactive lesions detected using the AT8 antibody (pSer202/pThr205 tau) in adjacent tissue sections. The other CTE cases had low frequencies of cortical tau-immunoreactive deposits and no well-defined autoradiography binding. In-vitro 3H-MK-6240 binding studies to CTE brain homogenates in the case with autoradiography signal indicated high binding affinity (KD = 2.0 ± 0.9 nM, Bmax = 97 ± 24 nM, n = 3). All NFL players had negative Aβ-PET. There was variable, low-to-intermediate intensity 18F-MK-6240 uptake across participants: 16 had no cortical signal, 7 had medial temporal lobe (MTL) uptake, 2 had frontal uptake, and 4 had MTL and frontal uptake. NFL players had higher SUVr in the entorhinal cortex (d = 0.86, p = 0.001), and the parahippocampal gyrus (d = 0.39, p = 0.08). Voxelwise regressions showed increased uptake in NFL players in two bilateral anterior MTL clusters (p < 0.05 FWE). Higher parahippocampal and frontal-temporal SUVrs correlated with worse memory (r = -0.38, r = -0.40) and semantic fluency (r = -0.38, r = -0.48), respectively. CONCLUSION We present evidence of 3H-MK-6240 in-vitro binding to post-mortem CTE tissue homogenates and in vivo 18F-MK-6240 PET binding in the MTL among a subset of participants. Additional studies in larger samples and PET-to-autopsy correlations are required to further elucidate the potential of 18F-MK-6240 to detect tau pathology in CTE.
Collapse
Affiliation(s)
- Michael L Alosco
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston Medical Center, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jhony Mejía Pérez
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Julia E Culhane
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ranjani Shankar
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Nidhi Mundada
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Karen Smith
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Alinda Amuiri
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Breton Asken
- Department of Clinical & Health Psychology, 1Florida Alzheimer's Disease Research Center, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Jenna R Groh
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Annalise Miner
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Erika Pettway
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sydney Mosaheb
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Charles Windon
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Gustavo Mercier
- Molecular Imaging and Nuclear Medicine, Boston Medical Center, Boston, MA, USA
| | - Robert A Stern
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lea T Grinberg
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - David N Soleimani-Meigooni
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Thor D Stein
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S.Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Psychiatry and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ann C McKee
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S.Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Psychiatry and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric E Abrahamson
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
- School of Medicine and Public Health, Wisconsin Alzheimer's Institute, University of Wisconsin-Madison, Madison, USA
| | - Jesse Mez
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Renaud La Joie
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Daniel Schonhaut
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
- University of California, San Francisco (UCSF), Memory and Aging Center MC: 1207, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA.
| |
Collapse
|
17
|
Ioannou K, Bucci M, Tzortzakakis A, Savitcheva I, Nordberg A, Chiotis K. Tau PET positivity predicts clinically relevant cognitive decline driven by Alzheimer's disease compared to comorbid cases; proof of concept in the ADNI study. Mol Psychiatry 2025; 30:587-599. [PMID: 39179903 PMCID: PMC11746147 DOI: 10.1038/s41380-024-02672-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 08/26/2024]
Abstract
β-amyloid (Aβ) pathology is not always coupled with Alzheimer's disease (AD) relevant cognitive decline. We assessed the accuracy of tau PET to identify Aβ(+) individuals who show prospective disease progression. 396 cognitively unimpaired and impaired individuals with baseline Aβ and tau PET and a follow-up of ≥ 2 years were selected from the Alzheimer's Disease Neuroimaging Initiative dataset. The participants were dichotomously grouped based on either clinical conversion (i.e., change of diagnosis) or cognitive deterioration (fast (FDs) vs. slow decliners (SDs)) using data-driven clustering of the individual annual rates of cognitive decline. To assess cognitive decline in individuals with isolated Aβ(+) or absence of both Aβ and tau (T) pathologies, we investigated the prevalence of non-AD comorbidities and FDG PET hypometabolism patterns suggestive of AD. Baseline tau PET uptake was higher in Aβ(+)FDs than in Aβ(-)FD/SDs and Aβ(+)SDs, independently of baseline cognitive status. Baseline tau PET uptake identified MCI Aβ(+) Converters and Aβ(+)FDs with an area under the curve of 0.85 and 0.87 (composite temporal region of interest) respectively, and was linearly related to the annual rate of cognitive decline in Aβ(+) individuals. The T(+) individuals constituted largely a subgroup of those being Aβ(+) and those clustered as FDs. The most common biomarker profiles in FDs (n = 70) were Aβ(+)T(+) (n = 34, 49%) and Aβ(+)T(-) (n = 19, 27%). Baseline Aβ load was higher in Aβ(+)T(+)FDs (M = 83.03 ± 31.42CL) than in Aβ(+)T(-)FDs (M = 63.67 ± 26.75CL) (p-value = 0.038). Depression diagnosis was more prevalent in Aβ(+)T(-)FDs compared to Aβ(+)T(+)FDs (47% vs. 15%, p-value = 0.021), as were FDG PET hypometabolism pattern not suggestive of AD (86% vs. 50%, p-value = 0.039). Our findings suggest that high tau PET uptake is coupled with both Aβ pathology and accelerated cognitive decline. In cases of isolated Aβ(+), cognitive decline may be associated with changes within the AD spectrum in a multi-morbidity context, i.e., mixed AD.
Collapse
Affiliation(s)
- Konstantinos Ioannou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Marco Bucci
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Antonios Tzortzakakis
- Division of Radiology, Department for Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Medical Radiation Physics and Nuclear Medicine, Section for Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Irina Savitcheva
- Medical Radiation Physics and Nuclear Medicine, Section for Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
18
|
Ghaderi S, Mohammadi S, Fatehi F. Diamagnetic Signature of Beta-Amyloid (Aβ) and Tau (τ) Tangle Pathology in Alzheimer's Disease: A Review. Aging Med (Milton) 2025; 8:e70006. [PMID: 39949469 PMCID: PMC11817029 DOI: 10.1002/agm2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/18/2024] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
The complex interplay between diamagnetic and paramagnetic substances within the brain, particularly in the context of Alzheimer's disease (AD), offers a rich landscape for investigation using advanced quantitative neuroimaging techniques. Although conventional approaches have focused on the paramagnetic properties of iron, emerging and promising research has highlighted the significance of diamagnetic signatures associated with beta-amyloid (Aβ) plaques and Tau (τ) protein aggregates. Quantitative susceptibility mapping (QSM) is a complex post-processing technique that visualizes and characterizes these subtle alterations in brain border tissue composition, such as the gray-white matter interface. Through voxel-wise separation of the contributions of diamagnetic and paramagnetic sources, QSM enabled the identification and quantification of Aβ and τ aggregates, even in the presence of iron. However, several challenges remain in utilizing diamagnetic signatures of Aβ and τ for clinical applications. These include the relatively small magnitude of the diamagnetic signal compared to paramagnetic iron, the need for high-resolution imaging and sophisticated analysis techniques, and the standardization of QSM acquisition and analysis protocols. Further research is necessary to refine QSM techniques, optimize acquisition parameters, and develop robust analysis pipelines to improve the sensitivity and specificity of detecting the diamagnetic nature of Aβ and τ aggregates. As our understanding of the diamagnetic properties of Aβ and τ continues to evolve, QSM is expected to play a pivotal role in advancing our knowledge of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Neurology DepartmentUniversity Hospitals of Leicester NHS TrustLeicesterUK
| |
Collapse
|
19
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated Appropriate Use Criteria for Amyloid and Tau PET: A Report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. J Nucl Med 2025:jnumed.124.268756. [PMID: 39778970 DOI: 10.2967/jnumed.124.268756] [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: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 01/11/2025] Open
Abstract
The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. Methods: The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. Results: For amyloid PET, 7 scenarios were rated as appropriate, 2 as uncertain, and 8 as rarely appropriate. For tau PET, 5 scenarios were rated as appropriate, 6 as uncertain, and 6 as rarely appropriate. Conclusion: AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Gil D Rabinovici
- Department of Neurology and Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California;
| | - David S Knopman
- Mayo Clinic Neurology and Neurosurgery, Rochester, Minnesota
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra Clinic, Pamplona, Spain
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri; Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Kevin J Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Peter Herscovitch
- Positron Emission Tomography Department, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Phillip H Kuo
- Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Jennifer H Lingler
- Department of Health and Community Systems, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | | | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Stephen P Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine, Brown University, Providence, Rhode Island
- Butler Hospital Memory and Aging Program, Providence, Rhode Island
| | | | | | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Molecular Neuroimaging, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
20
|
Appleton J, Finn Q, Zanotti-Fregonara P, Yu M, Faridar A, Nakawah MO, Zarate C, Carrillo MC, Dickerson BC, Rabinovici GD, Apostolova LG, Masdeu JC, Pascual B. Brain inflammation co-localizes highly with tau in mild cognitive impairment due to early-onset Alzheimer's disease. Brain 2025; 148:119-132. [PMID: 39013020 PMCID: PMC11706285 DOI: 10.1093/brain/awae234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Brain inflammation, with an increased density of microglia and macrophages, is an important component of Alzheimer's disease and a potential therapeutic target. However, it is incompletely characterized, particularly in patients whose disease begins before the age of 65 years and, thus, have few co-pathologies. Inflammation has been usefully imaged with translocator protein (TSPO) PET, but most inflammation PET tracers cannot image subjects with a low-binder TSPO rs6971 genotype. In an important development, participants with any TSPO genotype can be imaged with a novel tracer, 11C-ER176, that has a high binding potential and a more favourable metabolite profile than other TSPO tracers currently available. We applied 11C-ER176 to detect brain inflammation in mild cognitive impairment (MCI) caused by early-onset Alzheimer's disease. Furthermore, we sought to correlate the brain localization of inflammation, volume loss, elevated amyloid-β (Aβ)and tau. We studied brain inflammation in 25 patients with early-onset amnestic MCI (average age 59 ± 4.5 years, 10 female) and 23 healthy controls (average age 65 ± 6.0 years, 12 female), both groups with a similar proportion of all three TSPO-binding affinities. 11C-ER176 total distribution volume (VT), obtained with an arterial input function, was compared across patients and controls using voxel-wise and region-wise analyses. In addition to inflammation PET, most MCI patients had Aβ (n = 23) and tau PET (n = 21). For Aβ and tau tracers, standard uptake value ratios were calculated using cerebellar grey matter as region of reference. Regional correlations among the three tracers were determined. Data were corrected for partial volume effect. Cognitive performance was studied with standard neuropsychological tools. In MCI caused by early-onset Alzheimer's disease, there was inflammation in the default network, reaching statistical significance in precuneus and lateral temporal and parietal association cortex bilaterally, and in the right amygdala. Topographically, inflammation co-localized most strongly with tau (r = 0.63 ± 0.24). This correlation was higher than the co-localization of Aβ with tau (r = 0.55 ± 0.25) and of inflammation with Aβ (0.43 ± 0.22). Inflammation co-localized least with atrophy (-0.29 ± 0.26). These regional correlations could be detected in participants with any of the three rs6971 TSPO polymorphisms. Inflammation in Alzheimer's disease-related regions correlated with impaired cognitive scores. Our data highlight the importance of inflammation, a potential therapeutic target, in the Alzheimer's disease process. Furthermore, they support the notion that, as shown in experimental tissue and animal models, the propagation of tau in humans is associated with brain inflammation.
Collapse
Affiliation(s)
- Johanna Appleton
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Quentin Finn
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | | | - Meixiang Yu
- Cyclotron and Radiopharmaceutical Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Alireza Faridar
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Mohammad O Nakawah
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Carlos Zarate
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Maria C Carrillo
- Medical & Scientific Relations Division, Alzheimer's Association, Chicago, IL 60603, USA
| | | | - Gil D Rabinovici
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|
21
|
Khalighi MM, Young CB, Weiss S, Zeineh M, Davidzon G, Mormino E, Zaharchuk G. Enhancing the Diagnostic Accuracy of Amyloid PET: The Impact of MR-Guided PET Reconstruction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.04.25319996. [PMID: 39802766 PMCID: PMC11722500 DOI: 10.1101/2025.01.04.25319996] [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] [Indexed: 01/22/2025]
Abstract
18F-Florbetaben (FBB) uptake in the supratentorial cortex is indicative of amyloid positivity. Due to PET's low spatial resolution, image noise, and spill-over of signal from adjacent white-matter into gray-matter, there are inconsistencies in ratings among trained readers. A set of 264 18F-Florbetaben (amyloid) PET/MRI exams were reconstructed using conventional ordered subset expectation maximization (OSEM) method and MR-guided block sequential regularized expectation maximization (MRgBSREM) method. Images from 264 patients reconstructed by OSEM method and rated by 3 trained readers. Fifty-three exams were rated inconsistently and were mixed with another 53 exams which were rated consistently. These 106 subjects were then rated by our readers using the MRgBSREM PET reconstruction method. Centiloids (CL) were measured using both reconstruction methods. Signal to noise ratio (SNR) was calculated in frontal, anterior/posterior cingulate, lateral parietal, and lateral temporal regions for both reconstruction methods. There is significant correlation between CL measured by OSEM and MRgBSREM methods with R2=0.99. MRgBSREM enhanced the SNR in all regions by average of 21%. The number of inconsistent exams dropped by 64% using MRgBSREM method as compared with OSEM method. Using Fleiss-Kappa statistical test, the agreement between readers was raised from "Fair" to "Significant" in the 106-subjects subset. PET reconstruction with MR priors can significantly improve the consistency of ratings among trained readers. Given the prevalence of inconsistent ratings in amyloid PET, methods that enhance the ability to distinguish intermediate amyloid levels could be valuable for the widespread adoption of this modality.
Collapse
|
22
|
Dickerson BC, Atri A, Clevenger C, Karlawish J, Knopman D, Lin P, Norman M, Onyike C, Sano M, Scanland S, Carrillo M. The Alzheimer's Association clinical practice guideline for the Diagnostic Evaluation, Testing, Counseling, and Disclosure of Suspected Alzheimer's Disease and Related Disorders (DETeCD-ADRD): Executive summary of recommendations for specialty care. Alzheimers Dement 2025; 21:e14337. [PMID: 39713957 PMCID: PMC11772716 DOI: 10.1002/alz.14337] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 12/24/2024]
Abstract
US clinical practice guidelines for the diagnostic evaluation of cognitive impairment due to Alzheimer's disease (AD) or a related dementia (ADRD) are two decades old. This evidence-based guideline was developed to empower all clinicians to implement a structured approach for evaluating a patient with symptoms that may represent clinical AD/ADRD. An expert workgroup conducted a review of 7374 publications (133 met inclusion criteria) and developed recommendations as steps in an evaluation process. This summary briefly reviews core recommendations and details specialist recommendations of a high-quality, evidence-supported evaluation process aimed at characterizing, diagnosing, and disclosing the patient's cognitive functional status, cognitive-behavioral syndrome, and likely underlying brain disease so that optimal care plans to maximize patient/care partner dyad quality of life can be developed; a companion article summarizes primary care recommendations. If clinicians use the recommendations in this guideline and health-care systems provide adequate resources, outcomes should improve in most patients in most practice settings. HIGHLIGHTS: US clinical practice guidelines for the diagnostic evaluation of cognitive impairment due to Alzheimer's disease (AD) or related dementias (ADRD) are decades old and aimed at specialists. This evidence-based guideline was developed to empower all-including primary care-clinicians to implement a structured approach for evaluating a patient with symptoms that may represent clinical AD/ADRD. This summary focuses on recommendations appropriate for specialty practice settings, forming key elements of a high-quality, evidence-supported evaluation process aimed at characterizing, diagnosing, and disclosing the patient's cognitive functional status, cognitive-behavioral syndrome, and likely underlying brain disease so that optimal care plans to maximize patient/care partner dyad quality of life can be developed; a companion article summarizes primary care recommendations. If clinicians use this guideline and health-care systems provide adequate resources, outcomes should improve in most patients in most practice settings.
Collapse
Affiliation(s)
- Bradford C. Dickerson
- Frontotemporal Disorders Unit, Department of NeurologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Alireza Atri
- Banner Sun Health Research Institute and Banner Alzheimer's InstituteSun CityArizonaUSA
- Department of NeurologyCenter for Brain/Mind MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Carolyn Clevenger
- Department of Neurology, Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Jason Karlawish
- Departments of Medicine, Medical Ethics and Health Policy, and Neurology, Perelman School of Medicine, Penn Memory CenterUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David Knopman
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Pei‐Jung Lin
- Center for the Evaluation of Value and Risk in HealthInstitute for Clinical Research and Health Policy Studies, Tufts Medical CenterBostonMassachusettsUSA
| | - Mary Norman
- Cedars‐Sinai Medical CenterCulver CityCaliforniaUSA
| | - Chiadi Onyike
- Division of Geriatric Psychiatry and NeuropsychiatryThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mary Sano
- James J. Peters VAMCBronxNew YorkUSA
- Department of PsychiatryAlzheimer's Disease Research CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Maria Carrillo
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
23
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated appropriate use criteria for amyloid and tau PET: A report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. Alzheimers Dement 2025; 21:e14338. [PMID: 39776249 PMCID: PMC11772739 DOI: 10.1002/alz.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. METHODS The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. RESULTS For amyloid PET, seven scenarios were rated as appropriate, two as uncertain, and eight as rarely appropriate. For tau PET, five scenarios were rated as appropriate, six as uncertain, and six as rarely appropriate. DISCUSSION AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease. HIGHLIGHTS A multidisciplinary workgroup convened by the Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging updated the appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. The goal of these updated AUC is to assist clinicians in identifying clinical scenarios in which amyloid or tau PET may be useful for guiding the diagnosis and management of patients who have, or are at risk for, cognitive decline These updated AUC are intended for dementia specialists who spend a significant proportion of their clinical effort caring for patients with cognitive complaints, as well as serve as a general reference for a broader audience interested in implementation of amyloid and tau PET in clinical practice.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Department of Neurology and Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Javier Arbizu
- Department of Nuclear MedicineUniversity of Navarra ClinicPamplonaSpain
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer's Disease Research CenterWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Kevin J. Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Oskar Hansson
- Department of Clinical Sciences MalmöClinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory Clinic, Skåne University HospitalSkånes universitetssjukhusMalmöSweden
| | - Peter Herscovitch
- Positron Emission Tomography DepartmentNational Institutes of Health Clinical CenterBethesdaMarylandUSA
| | - Phillip H. Kuo
- Medical Imaging, Medicine, and Biomedical EngineeringUniversity of ArizonaTucsonArizonaUSA
| | - Jennifer H. Lingler
- Department of Health and Community SystemsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Satoshi Minoshima
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUtahUSA
| | | | - Julie C. Price
- Department of RadiologyMassachusetts General Hospital, BostonCharlestownMassachusettsUSA
| | - Stephen P. Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine at Brown UniversityProvidenceRhode IslandUSA
- Butler Hospital Memory and Aging ProgramProvidenceRhode IslandUSA
| | | | - Maria C. Carrillo
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Keith A. Johnson
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Molecular Neuroimaging, Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Departments of Neurology and RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
24
|
Sagar S, Khan D, Kumar R. PET-Computed Tomography-MR Imaging in Central Nervous System Disorders with Cognitive and Motor Impairment. PET Clin 2025; 20:101-111. [PMID: 39477721 DOI: 10.1016/j.cpet.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Neuroimaging, particularly positron emission tomography (PET), plays a crucial role in diagnosing and managing brain disorders by providing insights into diverse neuropathologies such as vascular issues, infections, inflammation, degenerative diseases, and tumors. In dementia, [18F]FDG-PET helps predict Alzheimer's disease (AD) development from mild cognitive impairment, revealing metabolic reductions in specific brain regions. PET's evolution with novel radiotracers and advanced imaging techniques addresses diagnostic challenges and enhances disease monitoring. Despite limitations like off-target binding, PET remains indispensable in clinical neurology, offering noninvasive insights into brain functions, disease progression, and treatment responses.
Collapse
Affiliation(s)
- Sambit Sagar
- Diagnostic Nuclear Medicine Division, Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Dikhra Khan
- Diagnostic Nuclear Medicine Division, Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Rakesh Kumar
- Diagnostic Nuclear Medicine Division, Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, Delhi, India.
| |
Collapse
|
25
|
Mormino EC, Biber SA, Rahman‐Filipiak A, Arfanakis K, Clark L, Dage JL, Detre JA, Dickerson BC, Donohue MC, Kecskemeti S, Hohman TJ, Jagust WJ, Keene DC, Kukull W, Levendovszky SR, Rosen H, Thompson PM, Villemagne VL, Wolk DA, Okonkwo OC, Rabinvovici GD, Rivera‐Mindt M, Foroud T, Johnson SC. The Consortium for Clarity in ADRD Research Through Imaging (CLARiTI). Alzheimers Dement 2025; 21:e14383. [PMID: 39588767 PMCID: PMC11772703 DOI: 10.1002/alz.14383] [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: 06/07/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/27/2024]
Abstract
The presence of multiple pathologies is the largest predictor of dementia. A major gap in the field is the in vivo detection of mixed pathologies and their antecedents. The Alzheimer's Disease Research Centers (ADRCs) are uniquely positioned to address this gap. The ADRCs longitudinally follow ≈ 17,000 participants, ranging from cognitively unimpaired to dementia, arising from Alzheimer's disease (AD) and related dementias (ADRD; e.g., AD, Lewy body disorders, vascular). Motivated by the Alzheimer's Disease Neuroimaging Initiative's (ADNI) impact, the ADRC Consortium for Clarity in ADRD Research Through Imaging (CLARiTI) was formed. Leveraging existing ADRC infrastructure, CLARiTI will integrate standardized imaging and plasma collection to characterize mixed pathologies and use community-engaged research methods to ensure that ≥ 25% of the sample is from underrepresented populations (e.g., ethnoculturally minoritized, low education). The resulting ADRD profiles, within a more diverse sample, will provide key resources for ADRCs and an unprecedented, more generalizable publicly available imaging-plasma dataset. HIGHLIGHTS: In vivo detection of mixed pathologies is critical for Alzheimer's disease and related dementias research. The Alzheimer's Disease Research Centers (ADRCs) are uniquely positioned to address gaps related to mixed pathologies. The ADRC Consortium for Clarity in ADRD Research Through Imaging (CLARiTI) will enhance this national program by adding standardized imaging and plasma collection to existing ADRC infrastructure. This effort will provide key resources for ADRCs and an unprecedented publicly available imaging-plasma-neuropath dataset.
Collapse
Affiliation(s)
- Elizabeth C. Mormino
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of Medicine, Cogen FacilityStanfordCaliforniaUSA
| | - Sarah A. Biber
- National Alzheimer's Coordinating CenterUniversity of WashingtonSeattleWashingtonUSA
| | | | | | - Lindsay Clark
- Department of MedicineUniversity of Wisconsin School of Medicine and Public Health, Health Sciences Learning CenterMadisonWisconsinUSA
| | - Jeffrey L. Dage
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - John A. Detre
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Bradford C. Dickerson
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Michael C. Donohue
- Department of NeurologyKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Alzheimer's Therapeutic Research Institute (ATRI), University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Steven Kecskemeti
- Department of MedicineUniversity of Wisconsin School of Medicine and Public Health, Health Sciences Learning CenterMadisonWisconsinUSA
| | - Timothy J. Hohman
- Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - William J. Jagust
- Department of EpidemiologySchool of Public HealthUniversity of CaliforniaBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Dirk C. Keene
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Walter Kukull
- National Alzheimer's Coordinating CenterUniversity of WashingtonSeattleWashingtonUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | | | - Howie Rosen
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Paul M. Thompson
- Department of Ophthalmology, Psychiatry and the Behavioral Sciences, RadiologyPsychiatry, and Engineering, Keck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - David A. Wolk
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ozioma C. Okonkwo
- Department of MedicineUniversity of Wisconsin School of Medicine and Public Health, Health Sciences Learning CenterMadisonWisconsinUSA
| | - Gil D. Rabinvovici
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Monica Rivera‐Mindt
- Department of PsychologyFordham UniversityBronxNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sterling C. Johnson
- Department of MedicineUniversity of Wisconsin School of Medicine and Public Health, Health Sciences Learning CenterMadisonWisconsinUSA
| |
Collapse
|
26
|
Shi L, Wang X, Si H, Song W. PDE4D inhibitors: Opening a new era of PET diagnostics for Alzheimer's disease. Neurochem Int 2025; 182:105903. [PMID: 39647702 DOI: 10.1016/j.neuint.2024.105903] [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: 04/23/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024]
Abstract
As the incidence of Alzheimer's disease (AD) continues to rise, the need for an effective PET radiotracer to facilitate early diagnosis has become more pressing than ever before in modern medicine. Phosphodiesterase (PDE) is closely related to cognitive impairment and neuroinflammatory processes in AD. Current research progress shows that specific PDE4D inhibitors radioligands can bind specifically to the PDE4D enzyme in the brain, thereby showing pathology-related signal enhancement in AD animal models, indicating the potential of these ligands as effective radiotracers. At the same time, we need to pay attention to the important role computer aided drug design (CADD) plays in advancing AD drug design and PET imaging. Future research will verify the potential of these ligands in clinical applications through computer simulation techniques, providing patients with timely intervention and treatment, which is of great significance.
Collapse
Affiliation(s)
- Luyang Shi
- College of Life Science, Qingdao University, Qingdao, China
| | - Xue Wang
- College of Life Science, Qingdao University, Qingdao, China
| | - Hongzong Si
- Laboratory of New Fibrous Materials and Modern Textile, The State Key Laboratory, Qingdao University, Qingdao, China.
| | - Wangdi Song
- School of Chemistry and Chemical Engineering, Shihezi University, Shihezi, China
| |
Collapse
|
27
|
Juengling F, Wuest F, Schirrmacher R, Abele J, Thiel A, Soucy JP, Camicioli R, Garibotto V. PET Imaging in Dementia: Mini-Review and Canadian Perspective for Clinical Use. Can J Neurol Sci 2025; 52:26-38. [PMID: 38433571 DOI: 10.1017/cjn.2024.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
PET imaging is increasingly recognized as an important diagnostic tool to investigate patients with cognitive disturbances of possible neurodegenerative origin. PET with 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), assessing glucose metabolism, provides a measure of neurodegeneration and allows a precise differential diagnosis among the most common neurodegenerative diseases, such as Alzheimer's disease, frontotemporal dementia or dementia with Lewy bodies. PET tracers specific for the pathological deposits characteristic of different neurodegenerative processes, namely amyloid and tau deposits typical of Alzheimer's Disease, allow the visualization of these aggregates in vivo. [18F]FDG and amyloid PET imaging have reached a high level of clinical validity and are since 2022 investigations that can be offered to patients in standard clinical care in most of Canada.This article will briefly review and summarize the current knowledge on these diagnostic tools, their integration into diagnostic algorithms as well as perspectives for future developments.
Collapse
Affiliation(s)
- Freimut Juengling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Faculty, University of Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ralf Schirrmacher
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Jonathan Abele
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Department of Neurology and Neurosurgery, Lady Davis Institute for Medical Research, McGill University, Montréal, QC, Canada
| | - Jean-Paul Soucy
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Valentina Garibotto
- Diagnostic Department, Nuclear Medicine and Molecular Imaging Division, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
28
|
Bischof GN, Jaeger E, Giehl K, Jessen F, Onur OA, O'Bryant S, Kara E, Weiss PH, Drzezga A. Cortical Tau Aggregation Patterns Associated With Apraxia in Patients With Alzheimer Disease. Neurology 2024; 103:e210062. [PMID: 39626130 PMCID: PMC11614392 DOI: 10.1212/wnl.0000000000210062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/01/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Apraxia is a frequently observed symptom in Alzheimer disease (AD), but the causal pathomechanism underlying this dysfunction is not well understood. Previous studies have demonstrated associations between various cognitive dysfunctions in AD and cortical tau deposition in specific brain areas, suggesting a causal relationship. Thus, we hypothesized that specific regional patterns of tau pathology in praxis-related brain regions may be associated with apraxic deficits in AD. For this purpose, we performed PET imaging with the second-generation tau-PET tracer [18F]PI-2620 in a well-defined group of patients with AD (N = 33) who had been systematically assessed for apraxia. METHODS Patients with a biomarker-confirmed diagnosis of AD were recruited in addition to a sample of cognitively unimpaired (CU1) control participants. Both groups underwent apraxia assessments with the Dementia Apraxia Screening Test. In addition, PET imaging with [18F]PI-2620 was performed to assess tau pathology in the patients with AD. To specifically investigate the association of apraxia severity with regional tau pathology, we compared the PET data from this group with an independent sample of amyloid-negative cognitively intact participants (CU2) by generation of z-score deviation maps and submitted these maps to a voxel-based multiple regression analysis. RESULTS A total of 120 participants (39% female) with a mean age of 67.9 (9.2) years were included in the study (AD = 33; CU1; N = 33; CU2; N = 54). We identified a significant correlation between circumscribed clusters of tau aggregation in praxis-related brain regions (including parietal (angular gyrus), temporal, and occipital regions) and severity of apraxia in patients with AD. By contrast, no significant correlations between tau tracer uptake in primary motor cortex or subcortical brain regions and apraxia were observed. DISCUSSION These results suggest that tau deposition in specific cortical praxis-related brain regions may induce local neuronal dysfunction leading to a dose-dependent functional decline in praxis performance in AD. The awareness of this relationship could further refine a differentiated individual diagnostic characterization and classification of patients with AD.
Collapse
Affiliation(s)
- Gérard N Bischof
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Elena Jaeger
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Kathrin Giehl
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Frank Jessen
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Oezguer A Onur
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Sid O'Bryant
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Esra Kara
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Peter H Weiss
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Alexander Drzezga
- From the Multimodal Neuroimaging Group, Department of Nuclear Medicine (G.N.B., E.J., K.G., A.D.), Department of Psychiatry (F.J.), Department of Neurology (O.A.O., E.K., P.H.W.), Medical Faculty and University Hospital of Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine II, Research Center Juelich; German Center for Neurodegenerative Diseases (F.J.), Bonn/Cologne, Germany; Institute for Translational Research (S.O.B.), and Department of Family Medicine (S.O.B.), Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth; and Cognitive Neuroscience (P.H.W.), Institute for Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| |
Collapse
|
29
|
Kumar R, Waisberg E, Ong J, Paladugu P, Amiri D, Saintyl J, Yelamanchi J, Nahouraii R, Jagadeesan R, Tavakkoli A. Artificial Intelligence-Based Methodologies for Early Diagnostic Precision and Personalized Therapeutic Strategies in Neuro-Ophthalmic and Neurodegenerative Pathologies. Brain Sci 2024; 14:1266. [PMID: 39766465 PMCID: PMC11674895 DOI: 10.3390/brainsci14121266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Advancements in neuroimaging, particularly diffusion magnetic resonance imaging (MRI) techniques and molecular imaging with positron emission tomography (PET), have significantly enhanced the early detection of biomarkers in neurodegenerative and neuro-ophthalmic disorders. These include Alzheimer's disease, Parkinson's disease, multiple sclerosis, neuromyelitis optica, and myelin oligodendrocyte glycoprotein antibody disease. This review highlights the transformative role of advanced diffusion MRI techniques-Neurite Orientation Dispersion and Density Imaging and Diffusion Kurtosis Imaging-in identifying subtle microstructural changes in the brain and visual pathways that precede clinical symptoms. When integrated with artificial intelligence (AI) algorithms, these techniques achieve unprecedented diagnostic precision, facilitating early detection of neurodegeneration and inflammation. Additionally, next-generation PET tracers targeting misfolded proteins, such as tau and alpha-synuclein, along with inflammatory markers, enhance the visualization and quantification of pathological processes in vivo. Deep learning models, including convolutional neural networks and multimodal transformers, further improve diagnostic accuracy by integrating multimodal imaging data and predicting disease progression. Despite challenges such as technical variability, data privacy concerns, and regulatory barriers, the potential of AI-enhanced neuroimaging to revolutionize early diagnosis and personalized treatment in neurodegenerative and neuro-ophthalmic disorders is immense. This review underscores the importance of ongoing efforts to validate, standardize, and implement these technologies to maximize their clinical impact.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA; (R.K.); (J.S.)
| | - Ethan Waisberg
- Department of Clinical Neurosciences, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK;
| | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St, Ann Arbor, MI 48105, USA
| | - Phani Paladugu
- Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut St, Philadelphia, PA 19107, USA;
- Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Dylan Amiri
- Department of Biology, University of Miami, 1301 Memorial Dr, Coral Gables, FL 33146, USA;
- Mecklenburg Neurology Group, 3541 Randolph Rd #301, Charlotte, NC 28211, USA;
| | - Jeremy Saintyl
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA; (R.K.); (J.S.)
| | - Jahnavi Yelamanchi
- Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY 11201, USA;
| | - Robert Nahouraii
- Mecklenburg Neurology Group, 3541 Randolph Rd #301, Charlotte, NC 28211, USA;
| | - Ram Jagadeesan
- Whiting School of Engineering, Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA;
| | - Alireza Tavakkoli
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, 1664 N Virginia St, Reno, NV 89557, USA;
| |
Collapse
|
30
|
Gupta A, Tripathi M, Sharma V, Ravindra SG, Jain S, Madhu G, Anjali, Yadav J, Singh I, Rajan R, Vishnu VY, Patil V, Nehra A, Singh MB, Bhatia R, Sharma A, Srivastava AK, Gaikwad S, Tripathi M, Srivastava MVP. Utility of Tau PET in the diagnostic work up of neurodegenerative dementia among Indian patients. J Neurol Sci 2024; 467:123292. [PMID: 39550784 DOI: 10.1016/j.jns.2024.123292] [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: 06/12/2024] [Revised: 07/20/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND AND OBJECTIVES Tau PET is being increasingly appraised as a novel diagnostic modality for dementia work up. Given limited data among South Asians, we assessed the frequency, patterns, phenotypic associations and incremental value of positive Tau PET scans in clinically diagnosed neurodegenerative dementia. METHODS This cross-sectional study recruited consecutive patients of Alzheimer's disease (AD) and non-AD syndromes (September 2021 to October 2022, India). Participants underwent clinical interview, cognitive assessment, MRI brain and tau PET scan ([F-18]ML-104). Visual read in a priori regions of interest was used to identify patterns of tau deposition in the brain. RESULTS We recruited 54 participants (mean age: 63.2 ± 9.2 years, 64.8 % men, 77.8 % dementia, 70.4 % early onset cases, 37.8 % APOE4+). The analysis identified abnormal tau uptake in 40/54 (74.1 %) participants; with uptake in AD signature areas in 27/40 (67.5 %) cases [cortical subtype (74.1 %), limbic (14.8 %), combined cortical/limbic (11.1 %)], and patterns not conforming to AD in 13/40 (32.5 %) cases. Tau PET substantiated the diagnosis of AD among 17/19 (89.5 %) cases with clinically diagnosed AD dementia, 8/23 (34.8 %) cases with suspected non-AD cause, and 2/12 (16.7 %) cases with mild cognitive impairment. A trend for increasing proportion of early onset cases, and worsening cognition, behavior and functional ability was seen, from 'limbic' to 'combined cortical/limbic' to 'cortical' subgroups. CONCLUSION Tau PET is a useful modality to differentiate AD dementia from other neurodegenerative causes in the Indian setting where amyloid biomarkers are not widely available. Biological subtypes of AD map well onto clinical phenotypes and need study in larger cohorts.
Collapse
Affiliation(s)
- Anu Gupta
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India.
| | - Madhavi Tripathi
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Varuna Sharma
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Shubha G Ravindra
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Savyasachi Jain
- Department of Neuroimaging & Intervention Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Gifty Madhu
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | - Anjali
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Jyoti Yadav
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Inder Singh
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Roopa Rajan
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Venugopalan Y Vishnu
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Vaibhav Patil
- Department of Psychiatry, All India Institute of Medical Sciences, New Delhi, India
| | - Ashima Nehra
- Department of Clinical Neuropsychology, All India Institute of Medical Sciences, New Delhi, India
| | - Mamta Bhushan Singh
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Rohit Bhatia
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Achal K Srivastava
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Shailesh Gaikwad
- Department of Neuroimaging & Intervention Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - M V Padma Srivastava
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
31
|
Ikari Y, Akamatsu G, Matsumoto K, Yamane T, Senda M, Fukuchi K. Improved Correlation of 18F-Flortaucipir PET SUVRs and Clinical Stages in the Alzheimer Disease Continuum with the MUBADA/PERSI-Based Analysis. J Nucl Med Technol 2024; 52:340-347. [PMID: 38627012 DOI: 10.2967/jnmt.123.267113] [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: 12/28/2023] [Accepted: 02/02/2024] [Indexed: 12/06/2024] Open
Abstract
The Alzheimer disease (AD) continuum is a neurodegenerative disorder with cognitive decline and pathologic changes. Tau PET imaging can detect tau pathology, and 18F-flortaucipir PET imaging is expected to visualize progression through the stages of AD, for which quantitative assessment is essential. Two measurement methods, statistically defined multiblock barycentric discriminant analysis (MUBADA)/parametric estimation of reference signal intensity (PERSI) and anatomically defined tau meta-volume of interest (VOI)/cerebellar gray matter (CGM) for SUV ratio (SUVR), were compared in this study to assess their relationship to AD clinical stage using 2 open multicenter PET databases. Methods: Data were selected for 106 cases from 2 databases, AMED Preclinical AD study (AMED-PRE) (n = 15) and Alzheimer Disease Neuroimaging Initiative 3 (n = 91). The data of the participants were categorized into 4 groups based on the clinical criteria. Tau PET imaging was conducted using 18F-flortaucipir, and the 2 SUVR measurement methods, MUBADA/PERSI and tau meta-VOI/CGM, were compared among different clinical categories: amyloid-negative cognitively normal, preclinical AD, amyloid-negative mild cognitive impairment (MCI), and amyloid-positive MCI. Results: Significant differences were found between cognitively normal and preclinical AD, as well as between cognitively normal and amyloid-positive MCI and between amyloid-negative MCI and -positive MCI in SUVR derived by MUBADA/PERSI, whereas SUVR by tau meta-VOI/CGM did not provide significant differences between any pair. The tau meta-VOI/CGM method consistently provided higher SUVRs and larger individual variations than MUBADA/PERSI, with a mean SUVR difference of 0.136 for the studied databases. Conclusion: MUBADA/PERSI provided the SUVR of 18F-flortaucipir uptake with better association with the clinical severity of the AD continuum and with smaller variability. The results support the usefulness of MUBADA/PERSI as a quantitative measure of 18F-flortaucipir uptake in multicenter studies using different PET systems and scanning methods. However, limitations of the study include the small sample size and the unbalanced distribution among clinical categories in the AMED Preclinical AD study database.
Collapse
Affiliation(s)
- Yasuhiko Ikari
- Department of Molecular Imaging Research, Kobe City Medical Center General Hospital, Kobe, Japan;
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Go Akamatsu
- Department of Molecular Imaging Research, Kobe City Medical Center General Hospital, Kobe, Japan
- National Institutes for Quantum Science and Technology, Chiba, Japan; and
| | - Keiichi Matsumoto
- Department of Molecular Imaging Research, Kobe City Medical Center General Hospital, Kobe, Japan
- Department of Radiological Technology, Faculty of Medical Science, Kyoto College of Medical Science, Nantan, Japan
| | - Tomohiko Yamane
- Department of Molecular Imaging Research, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Michio Senda
- Department of Molecular Imaging Research, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Kazuki Fukuchi
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
32
|
Lavrova A, Satoh R, Pham NTT, Nguyen A, Jack CR, Petersen RC, Ross RR, Dickson DW, Lowe VJ, Whitwell JL, Josephs KA. Investigating the feasibility of 18F-flortaucipir PET imaging in the antemortem diagnosis of primary age-related tauopathy (PART): An observational imaging-pathological study. Alzheimers Dement 2024; 20:8605-8614. [PMID: 39417408 DOI: 10.1002/alz.14301] [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: 06/06/2024] [Revised: 08/08/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Primary age-related tauopathy (PART) is characterized by neurofibrillary tangles and minimal β-amyloid deposition, diagnosed postmortem. This study investigates 18F-flortaucipir (FTP) PET imaging for antemortem PART diagnosis. METHODS We analyzed FTP PET scans from 50 autopsy-confirmed PART and 13 control subjects. Temporal lobe uptake was assessed both qualitatively and quantitatively. Demographic and clinicopathological characteristics and voxel-level uptake using SPM12 were compared between FTP-positive and FTP-negative cases. Intra-reader reproducibility was evaluated with Krippendorff's alpha. RESULTS Minimal/mild and moderate FTP uptake was seen in 32% of PART cases and 62% of controls, primarily in the left inferior temporal lobe. No demographic or clinicopathological differences were found between FTP-positive and FTP-negative cases. High intra-reader reproducibility (α = 0.83) was noted. DISCUSSION FTP PET imaging did not show a specific uptake pattern for PART diagnosis, indicating that in vivo PART identification using FTP PET is challenging. Similar uptake in controls suggests non-specific uptake in PART. HIGHLIGHTS 18F-flortaucipir (FTP) PET scans were analyzed for diagnosing PART antemortem. 32% of PART cases had minimal/mild FTP uptake in the left inferior temporal lobe. Similar to PART FTP uptake was found in 62% of control subjects. No specific uptake pattern was found, challenging in vivo PART diagnosis.
Collapse
Affiliation(s)
- Anna Lavrova
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryota Satoh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Aivi Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Reichard R Ross
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Pievani M, Ribaldi F, Toussas K, Da Costa S, Jorge J, Reynaud O, Chicherio C, Blouin JL, Scheffler M, Garibotto V, Jovicich J, Jelescu IO, Frisoni GB. Resting-state functional connectivity abnormalities in subjective cognitive decline: A 7T MRI study. Neurobiol Aging 2024; 144:104-113. [PMID: 39305703 DOI: 10.1016/j.neurobiolaging.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/23/2024] [Accepted: 09/11/2024] [Indexed: 10/21/2024]
Abstract
Resting-state functional connectivity (FC) MRI is sensitive to brain changes in Alzheimer's disease in preclinical stages, however studies in persons with subjective cognitive decline (SCD) have reported conflicting findings, and no study is available at 7T MRI. In this study, we investigated FC alterations in sixty-six participants recruited at the Geneva Memory Center (24 controls, 14 SCD, 28 cognitively impaired [CI]). Participants were classified as SCD if they reported cognitive complaints without objective cognitive deficits, and underwent 7T fMRI to assess FC in canonical brain networks and their association with cognitive/clinical features. SCD showed normal cognition, a trend for higher depressive symptoms, and normal AD biomarkers. Compared to the other two groups, SCD showed higher FC in frontal default mode network (DMN) and insular and superior temporal nodes of ventral attention network (VAN). Higher FC in the DMN and VAN was associated with worse cognition but not depression, suggesting that hyper-connectivity in these networks may be a signature of age-related cognitive decline in SCD at low risk of developing AD.
Collapse
Affiliation(s)
- M Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - F Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland; Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland
| | - K Toussas
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
| | - S Da Costa
- CIBM Center for Biomedical Imaging, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - J Jorge
- CSEM - Swiss Center for Electronics and Microtechnology, Bern, Switzerland
| | - O Reynaud
- CIBM Center for Biomedical Imaging, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Fondation Campus Biotech Geneva, Geneva, Switzerland
| | - C Chicherio
- Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland
| | - J L Blouin
- Genetic Medicine, Diagnostics Dept, University Hospitals and University of Geneva, Geneva, Switzerland
| | - M Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - V Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland; Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland; CIBM Center for Biomedical Imaging, Geneva, Switzerland
| | - J Jovicich
- Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - I O Jelescu
- CIBM Center for Biomedical Imaging, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Lausanne University Hospital (CHUV) and University of Lausanne (UNIL) Lausanne, Department of Radiology, Lausanne, Switzerland
| | - G B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland; Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
34
|
Simrén J, Ashton NJ, Suárez-Calvet M, Zetterberg H. Alzheimer's disease-Biomarkers, clinical evaluation or both? J Neuropsychol 2024. [PMID: 39543822 DOI: 10.1111/jnp.12401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Affiliation(s)
- Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, UCL, London, UK
- UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
35
|
Studart-Neto A, Barbosa BJAP, Coutinho AM, de Souza LC, Schilling LP, da Silva MNM, Castilhos RM, Bertolucci PHF, Borelli WV, Gomes HR, Fernandes GBP, Barbosa MT, Balthazar MLF, Frota NAF, Forlenza OV, Smid J, Brucki SMD, Caramelli P, Nitrini R, Engelhardt E, Resende EDPF. Guidelines for the use and interpretation of Alzheimer's disease biomarkers in clinical practice in Brazil: recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2024; 18:e2024C001. [PMID: 39534442 PMCID: PMC11556292 DOI: 10.1590/1980-5764-dn-2024-c001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, the diagnostic accuracy of Alzheimer's disease has been enhanced by the development of different types of biomarkers that indicate the presence of neuropathological processes. In addition to improving patient selection for clinical trials, biomarkers can assess the effects of new treatments on pathological processes. However, there is concern about the indiscriminate and poorly supported use of biomarkers, especially in asymptomatic individuals or those with subjective cognitive decline. Difficulties interpreting these tests, high costs, and unequal access make this scenario even more challenging in healthcare. This article presents the recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology (Departamento Científico de Neurologia Cognitiva e Envelhecimento da Academia Brasileira de Neurologia) regarding the rational use and interpretation of Alzheimer's disease biomarkers in clinical practice. The clinical diagnosis of cognitive-behavioral syndrome is recommended as the initial step to guide the request for biomarkers.
Collapse
Affiliation(s)
- Adalberto Studart-Neto
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Breno José Alencar Pires Barbosa
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Pernambuco, Hospital das Clínicas, Recife, Centro de Ciências Médicas, Recife PE, Brazil
- Universidade Federal de Pernambuco, Empresa Brasileira de Serviços Hospitalares, Hospital das Clínicas, Departamento de Neurologia, Recife PE, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Radiologia, Centro de Medicina Nuclear, Laboratório de Investigação Médica (LIM 43), São Paulo SP, Brazil
- Hospital Sírio-Libanês, Medicina Nuclear e Serviço de PET-CT, São Paulo SP, Brazil
| | - Leonardo Cruz de Souza
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Lucas Porcello Schilling
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil
| | - Mari Nilva Maia da Silva
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Nina Rodrigues, Serviço de Neuropsiquiatria, São Luís MA, Brazil
| | - Raphael Machado Castilhos
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Centro de Neurologia Cognitiva e Comportamental, Porto Alegre RS, Brazil
| | - Paulo Henrique Ferreira Bertolucci
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| | - Wyllians Vendramini Borelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre RS, Brazil
| | - Hélio Rodrigues Gomes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Laboratório de Líquido Cefalorraquidiano, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Laboratório de Investigação Médica (LIM 15), São Paulo SP, Brazil
- Departamento Científico de Líquido Cefalorraquiano, Academia Brasileira de Neurologia, São Paulo SP, Brazil
| | | | - Maira Tonidandel Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Marcio Luiz Figueredo Balthazar
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil
| | - Norberto Anízio Ferreira Frota
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil
- Universidade de Fortaleza, Fortaleza, CE, Brazil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil
| | - Jerusa Smid
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Paulo Caramelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Ricardo Nitrini
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Eliasz Engelhardt
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| |
Collapse
|
36
|
Li A, Zhao R, Zhang M, Sun P, Cai Y, Zhu L, Kung H, Han Y, Wang X, Guo T. [ 18F]-D3FSP β-amyloid PET imaging in older adults and alzheimer's disease. Eur J Nucl Med Mol Imaging 2024; 51:3990-4000. [PMID: 38976036 DOI: 10.1007/s00259-024-06835-2] [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: 12/06/2023] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
PURPOSE [18F]-D3FSP is a new β-amyloid (Aβ) PET imaging tracer designed to decrease nonspecific signals in the brain by reducing the formation of the N-demethylated product. However, its optimal reference region for calculating the standardized uptake value ratio (SUVR) and its relation to the well-established biomarkers of Alzheimer's disease (AD) are still unclear. METHODS We recruited 203 participants from the Greater Bay Area Healthy Aging Brain Study (GHABS) to undergo [18F]-D3FSP Aβ PET imaging. We analyzed plasma Aβ42/Aβ40, p-Tau181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) using the Simoa platform. We compared the standardized uptake value (SUV) of five reference regions (cerebellum, cerebellum cortex, brainstem/PONs, white matter, composite of the four regions above) and AD typical cortical region (COMPOSITE) SUVR among different clinical groups. The association of D3FSP SUVR with plasma biomarkers, imaging biomarkers, and cognition was also investigated. RESULTS Brainstem/PONs SUV showed the lowest fluctuation across diagnostic groups, and COMPOSITE D3FSP SUVR had an enormous effect distinguishing cognitively impaired (CI) individuals from cognitively unimpaired (CU) individuals. COMPOSITE SUVR (Referred to brainstem/PONs) was positively correlated with p-Tau181 (p < 0.001), GFAP (p < 0.001), NfL (p = 0.014) in plasma and temporal-metaROI tau deposition (p < 0.001), and negatively related to plasma Aβ42/Aβ40 (p < 0.001), temporal-metaROI cortical thickness (p < 0.01), residual hippocampal volume (p < 0.001) and cognition (p < 0.001). The voxel-wise analysis replicated these findings. CONCLUSION This study suggests brainstem/PONs as an optimal reference region for calculating D3FSP SUVR to quantify cortical Aβ plaques in the brain. [18F]-D3FSP could distinguish CI from CU and strongly correlates with well-established plasma biomarkers, tau PET, neurodegeneration, and cognitive decline. However, future head-to-head comparisons of [18F]-D3FSP PET images with other validated Aβ PET tracers or postmortem results are crucial.
Collapse
Affiliation(s)
- Anqi Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Ruiyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Mingkai Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Pan Sun
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Yue Cai
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Lin Zhu
- Beijing Normal University, Beijing, 100875, China
| | - Hank Kung
- University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ying Han
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China
- National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Xinlu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China.
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China.
| | - Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Institute of Biomedical Engineering, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China.
| |
Collapse
|
37
|
Vermeiren MR, Somsen J, Luurtsema G, Reesink FE, Verwey NA, Hempenius L, Tolboom N, Biessels GJ, Biesbroek JM, Vernooij MW, Veldhuijzen van Zanten SEM, Seelaar H, Coomans EM, Teunissen CE, Lemstra AW, van Harten AC, Visser LNC, van der Flier WM, van de Giessen E, Ossenkoppele R. The impact of tau-PET in a selected memory clinic cohort: rationale and design of the TAP-TAU study. Alzheimers Res Ther 2024; 16:230. [PMID: 39427210 PMCID: PMC11490118 DOI: 10.1186/s13195-024-01588-4] [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: 06/18/2024] [Accepted: 09/29/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Tau-PET is a diagnostic tool with high sensitivity and specificity for discriminating Alzheimer's disease (AD) dementia from other neurodegenerative disorders in well-controlled research environments. The role of tau-PET in real-world clinical practice, however, remains to be established. The aim of the TAP-TAU study is therefore to investigate the impact of tau-PET in clinical practice. METHODS TAP-TAU is a prospective, longitudinal multi-center study in 300 patients (≥ 50 years old) with mild cognitive impairment or mild dementia across five Dutch memory clinics. Patients are eligible if diagnostic certainty is < 85% after routine dementia screening and if the differential diagnosis includes AD. More specifically, we will include patients who (i) are suspected of having mixed pathology (e.g., AD and vascular pathology), (ii) have an atypical clinical presentation, and/or (iii) show conflicting or inconclusive outcomes on other tests (e.g., magnetic resonance imaging or cerebrospinal fluid). Participants will undergo a [18F]flortaucipir tau-PET scan, blood-based biomarker sampling, and fill out questionnaires on patient reported outcomes and experiences. The primary outcomes are change (pre- versus post- tau-PET) in diagnosis, diagnostic certainty, patient management and patient anxiety and uncertainty. Secondary outcome measures are head-to-head comparisons between tau-PET and less invasive and lower cost diagnostic tools such as novel blood-based biomarkers and artificial intelligence-based classifiers. RESULTS TAP-TAU has been approved by the Medical Ethics Committee of the Amsterdam UMC. The first participant is expected to be included in October 2024. CONCLUSIONS In TAP-TAU, we will investigate the added clinical value of tau-PET in a real-world clinical setting, including memory clinic patients with diagnostic uncertainty after routine work-up. Findings of our study may contribute to recommendations regarding which patients would benefit most from assessment with tau-PET. This study is timely in the dawning era of disease modifying treatments as an accurate etiological diagnosis becomes increasingly important. TRIAL REGISTRATION This trial is registered and authorized on December 21st, 2023 in EU Clinical Trials with registration number 2023-505430-10-00.
Collapse
Affiliation(s)
- Marie R Vermeiren
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands.
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands.
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands.
| | - Joost Somsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Fransje E Reesink
- Department of Neurology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Nicolaas A Verwey
- Department of Neurology, Medical Center Leeuwarden, Leeuwarden, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | | | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - J Matthijs Biesbroek
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Neurology, Diakonessenhuis Hospital, Utrecht, Netherlands
| | - Meike W Vernooij
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Harro Seelaar
- Department of Neurology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Emma M Coomans
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | | | - Afina W Lemstra
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Argonde C van Harten
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Leonie N C Visser
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Psychology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Epidemiology and Data Science, Amsterdam UMC, Amsterdam, Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands.
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands.
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
| |
Collapse
|
38
|
Jagust WJ, Koeppe RA, Rabinovici GD, Villemagne VL, Harrison TM, Landau SM, the Alzheimer's Disease Neuroimaging Initiative. The ADNI PET Core at 20. Alzheimers Dement 2024; 20:7340-7349. [PMID: 39108002 PMCID: PMC11485322 DOI: 10.1002/alz.14165] [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: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) PET Core has evolved over time, beginning with positron emission tomography (PET) imaging of a subsample of participants with [18F]fluorodeoxyglucose (FDG)-PET, adding tracers for measurement of β-amyloid, followed by tau tracers. This review examines the evolution of the ADNI PET Core, the novel aspects of PET imaging in each stage of ADNI, and gives an accounting of PET images available in the ADNI database. The ADNI PET Core has been and continues to be a rich resource that provides quantitative PET data and preprocessed PET images to the scientific community, allowing interrogation of both basic and clinically relevant questions. By standardizing methods across different PET scanners and multiple PET tracers, the Core has demonstrated the feasibility of large-scale, multi-center PET studies. Data managed and disseminated by the PET Core has been critical to defining pathophysiological models of Alzheimer's disease (AD) and helped to drive methods used in modern therapeutic trials. HIGHLIGHTS: The ADNI PET Core began with FDG-PET and now includes three amyloid and three tau PET ligands. The PET Core has standardized acquisition and analysis of multitracer PET images. The ADNI PET Core helped to develop methods that have facilitated clinical trials in AD.
Collapse
Affiliation(s)
- William J. Jagust
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Robert A. Koeppe
- Department of RadiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Gil D. Rabinovici
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | | | | - Susan M. Landau
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | |
Collapse
|
39
|
Vermeiren MR, Calandri IL, van der Flier WM, van de Giessen E, Ossenkoppele R. Survey among experts on the future role of tau-PET in clinical practice and trials. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70033. [PMID: 39583643 PMCID: PMC11582687 DOI: 10.1002/dad2.70033] [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: 07/03/2024] [Accepted: 10/02/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Recent advancements in Alzheimer's disease (AD) biomarker research and clinical trials prompt reflection on the value and consequently appropriate use of tau positron emission tomography (tau-PET) in the future. METHODS We conducted an online survey among dementia and PET experts worldwide to investigate the anticipated future role of tau-PET in clinical practice and trials. RESULTS Two hundred sixty-eight dementia experts, comprising 143 clinicians and 121 researchers, covering six continents participated. The vast majority (90%) fostered a positive attitude toward the added value of tau-PET in clinical practice, particularly for staging, diagnosing, monitoring, and prognostication in a cognitively impaired memory clinic population. Experts anticipated an important role for tau-PET for participant selection (76%-100%) and measuring endpoints (75%-97%), in both anti-amyloid and anti-tau drug trials. DISCUSSION Our global survey study shows that dementia experts envision an important role for tau-PET in the future, both in clinical practice and in drug trials, beyond current guidelines and practices. Highlights Dementia experts envision an important role for tau-PET in the future.Experts indicate that a tau-PET scan could influence patient management.Experts anticipate the utility of tau-PET for participant selection and endpoints in drug trials.There is a gap between the anticipated usefulness of tau-PET and current clinical practices.
Collapse
Affiliation(s)
- Marie R. Vermeiren
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceBrain ImagingAmsterdamThe Netherlands
| | - Ismael L. Calandri
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Department of Cognitive NeurologyFleniBuenos AiresArgentina
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
- Epidemiology and Data ScienceVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Elsmarieke van de Giessen
- Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceBrain ImagingAmsterdamThe Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, NeurologyVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamThe Netherlands
- Clinical Memory Research UnitLund UniversityLundSweden
| |
Collapse
|
40
|
Pichet Binette A, Gaiteri C, Wennström M, Kumar A, Hristovska I, Spotorno N, Salvadó G, Strandberg O, Mathys H, Tsai LH, Palmqvist S, Mattsson-Carlgren N, Janelidze S, Stomrud E, Vogel JW, Hansson O. Proteomic changes in Alzheimer's disease associated with progressive Aβ plaque and tau tangle pathologies. Nat Neurosci 2024; 27:1880-1891. [PMID: 39187705 PMCID: PMC11452344 DOI: 10.1038/s41593-024-01737-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 07/23/2024] [Indexed: 08/28/2024]
Abstract
Proteomics can shed light on the dynamic and multifaceted alterations in neurodegenerative disorders like Alzheimer's disease (AD). Combining radioligands measuring β-amyloid (Aβ) plaques and tau tangles with cerebrospinal fluid proteomics, we uncover molecular events mirroring different stages of AD pathology in living humans. We found 127 differentially abundant proteins (DAPs) across the AD spectrum. The strongest Aβ-related proteins were mainly expressed in glial cells and included SMOC1 and ITGAM. A dozen proteins linked to ATP metabolism and preferentially expressed in neurons were independently associated with tau tangle load and tau accumulation. Only 20% of the DAPs were also altered in other neurodegenerative diseases, underscoring AD's distinct proteome. Two co-expression modules related, respectively, to protein metabolism and microglial immune response encompassed most DAPs, with opposing, staggered trajectories along the AD continuum. We unveil protein signatures associated with Aβ and tau proteinopathy in vivo, offering insights into complex neural responses and potential biomarkers and therapeutics targeting different disease stages.
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| | - Chris Gaiteri
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
- Rush University Alzheimer's Disease Center, Rush University, Chicago, IL, USA
| | - Malin Wennström
- Cognitive Disorder Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Atul Kumar
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Ines Hristovska
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Hansruedi Mathys
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- University of Pittsburgh Brain Institute and Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Rush University Alzheimer's Disease Center, Rush University, Chicago, IL, USA
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Jacob W Vogel
- Department of Clinical Sciences Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
41
|
Jack CR, Graf A, Burnham SC, Doty EG, Moebius HJ, Montenigro P, Siemers E, Sink KM, Shaw LM, Hansen CT, Wildsmith KR, Mahinrad S, Carrillo MC, Weber CJ. Application of the revised criteria for diagnosis and staging of Alzheimer's disease: Drug development and clinical practice. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e70013. [PMID: 39748835 PMCID: PMC11694534 DOI: 10.1002/trc2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 01/04/2025]
Abstract
The newly proposed revised criteria for diagnosis and staging of Alzheimer's disease (AD) by the Alzheimer's Association (AA) Workgroup represent a significant milestone in the field. These criteria offer objective measures for diagnosing and staging biological AD, bridging the gap between research and clinical care. Although implementation feasibility may vary across regions and settings, improving the availability and accuracy of biomarkers, especially plasma biomarkers, is expected to enhance the applicability of these criteria in clinical practice. The Fall 2023 Alzheimer's Association Research Roundtable (AARR) meeting served as a forum for gathering industry perspectives and feedback on these revised criteria, ensuring that the new criteria inform research, clinical trial design, and clinical care. In this article, we outline a summary of the newly proposed "Revised Criteria for Diagnosis and Staging of AD: AA Workgroup" and provide highlights from the AARR meeting in fall 2023. Highlights The Alzheimer's Association Research Roundtable (AARR) convened leaders from industry, academia, and government, to review the Revised Criteria for Diagnosis and Staging of AD: AA Workgroup, and gather industry perspectives and feedback on these revised criteria before its publication.The newly proposed revised criteria for diagnosis and staging of Alzheimer's disease (AD) by the AA's Workgroup represent a significant milestone, offering objective measures for the biological and staging of AD and bridging the gap between research and clinical care.Improving the availability and accuracy of biomarkers, especially blood-based biomarkers (BBMs) is expected to improve clinical research and enhance the applicability of these criteria in clinical practice.
Collapse
Affiliation(s)
| | - Ana Graf
- Novartis Pharma AGBaselSwitzerland
| | | | | | | | | | | | | | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of Medicine University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | | | | | | | | |
Collapse
|
42
|
Freiburghaus T, Pawlik D, Oliveira Hauer K, Ossenkoppele R, Strandberg O, Leuzy A, Rittmo J, Tremblay C, Serrano GE, Pontecorvo MJ, Beach TG, Smith R, Hansson O. Association of in vivo retention of [ 18f] flortaucipir pet with tau neuropathology in corresponding brain regions. Acta Neuropathol 2024; 148:44. [PMID: 39297933 DOI: 10.1007/s00401-024-02801-2] [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: 06/14/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/21/2024]
Abstract
[18F]Flortaucipir is an FDA-approved tau-PET tracer that is increasingly utilized in clinical settings for the diagnosis of Alzheimer's disease. Still, a large-scale comparison of the in vivo PET uptake to quantitative post-mortem tau pathology and to other co-pathologies is lacking. Here, we examined the correlation between in vivo [18F]flortaucipir PET uptake and quantitative post-mortem tau pathology in corresponding brain regions from the AVID A16 end-of-life study (n = 63). All participants underwent [18F]flortaucipir PET scans prior to death, followed by a detailed post-mortem neuropathological examination using AT8 (tau) immunohistochemistry. Correlations between [18F]flortaucipir standardized uptake value ratios (SUVRs) and AT8 immunohistochemistry were assessed across 18 regions-of-interest (ROIs). To assess [18F]flortaucipir specificity and level of detection for tau pathology, correlations between [18F]flortaucipir SUVR and neuritic plaque score and TDP-43 stage were also computed and retention was further assessed in individuals with possible primary age-related tauopathy (PART), defined as Thal phase ≤ 2 and Braak stage I-IV. We found modest-to-strong correlations between in vivo [18F]flortaucipir SUVR and post-mortem tau pathology density in corresponding brain regions in all neocortical regions analyzed (rho-range = 0.61-0.79, p < 0.0001 for all). The detection threshold of [18F]flortaucipir PET was determined to be 0.85% of surface area affected by tau pathology in a temporal meta-ROI, and 0.15% in a larger cortical meta-ROI. No significant associations were found between [18F]flortaucipir SUVRs and post-mortem tau pathology in individuals with possible PART. Further, there was no correlation observed between [18F]flortaucipir and level of amyloid-β neuritic plaque load (rho-range = - 0.16-0.12; p = 0.48-0.61) or TDP-43 stage (rho-range = - 0.10 to - 0.30; p = 0.18-0.65). In conclusion, our in vivo vs post-mortem study shows that the in vivo [18F]flortaucipir PET signal primarily reflects tau pathology, also at relatively low densities of tau proteinopathy, and does not bind substantially to tau neurites in neuritic plaques or in individuals with PART.
Collapse
Affiliation(s)
- Tove Freiburghaus
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Daria Pawlik
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Kevin Oliveira Hauer
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jonathan Rittmo
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | | | | | | | | | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, 20502, Malmö, Sweden.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, 20502, Malmö, Sweden.
| |
Collapse
|
43
|
Moskal P, Baran J, Bass S, Choiński J, Chug N, Curceanu C, Czerwiński E, Dadgar M, Das M, Dulski K, Eliyan KV, Fronczewska K, Gajos A, Kacprzak K, Kajetanowicz M, Kaplanoglu T, Kapłon Ł, Klimaszewski K, Kobylecka M, Korcyl G, Kozik T, Krzemień W, Kubat K, Kumar D, Kunikowska J, Mączewska J, Migdał W, Moskal G, Mryka W, Niedźwiecki S, Parzych S, Del Rio EP, Raczyński L, Sharma S, Shivani S, Shopa RY, Silarski M, Skurzok M, Tayefi F, Ardebili KT, Tanty P, Wiślicki W, Królicki L, Stępień EŁ. Positronium image of the human brain in vivo. SCIENCE ADVANCES 2024; 10:eadp2840. [PMID: 39270027 PMCID: PMC11397496 DOI: 10.1126/sciadv.adp2840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024]
Abstract
Positronium is abundantly produced within the molecular voids of a patient's body during positron emission tomography (PET). Its properties dynamically respond to the submolecular architecture of the tissue and the partial pressure of oxygen. Current PET systems record only two annihilation photons and cannot provide information about the positronium lifetime. This study presents the in vivo images of positronium lifetime in a human, for a patient with a glioblastoma brain tumor, by using the dedicated Jagiellonian PET system enabling simultaneous detection of annihilation photons and prompt gamma emitted by a radionuclide. The prompt gamma provides information on the time of positronium formation. The photons from positronium annihilation are used to reconstruct the place and time of its decay. In the presented case study, the determined positron and positronium lifetimes in glioblastoma cells are shorter than those in salivary glands and those in healthy brain tissues, indicating that positronium imaging could be used to diagnose disease in vivo.
Collapse
Affiliation(s)
- Paweł Moskal
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Jakub Baran
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Steven Bass
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
- Kitzbühel Centre for Physics, Kitzbühel, Austria
| | | | - Neha Chug
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Catalina Curceanu
- INFN, Laboratori Nazionali di Frascati, Via E. Fermi 40, 00044 Frascati, Italy
| | - Eryk Czerwiński
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Meysam Dadgar
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Manish Das
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Kamil Dulski
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Kavya V Eliyan
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Katarzyna Fronczewska
- Department of Nuclear Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Aleksander Gajos
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Krzysztof Kacprzak
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Marcin Kajetanowicz
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Tevfik Kaplanoglu
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Łukasz Kapłon
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Konrad Klimaszewski
- Department of Complex Systems, National Centre for Nuclear Research, 05-400 Otwock-Świerk, Poland
| | - Małgorzata Kobylecka
- Department of Nuclear Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Grzegorz Korcyl
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Tomasz Kozik
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Wojciech Krzemień
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
- High Energy Department, National Centre for Nuclear Research, 05-400 Otwock-Świerk, Poland
| | - Karol Kubat
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Deepak Kumar
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Jolanta Kunikowska
- Department of Nuclear Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Joanna Mączewska
- Department of Nuclear Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Wojciech Migdał
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Gabriel Moskal
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
- Department of Chemical Technology, Faculty of Chemistry of the Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Wiktor Mryka
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Szymon Niedźwiecki
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Szymon Parzych
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Elena P Del Rio
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Lech Raczyński
- Department of Complex Systems, National Centre for Nuclear Research, 05-400 Otwock-Świerk, Poland
| | - Sushil Sharma
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Shivani Shivani
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Roman Y Shopa
- Department of Complex Systems, National Centre for Nuclear Research, 05-400 Otwock-Świerk, Poland
| | - Michał Silarski
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Magdalena Skurzok
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Faranak Tayefi
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Keyvan T Ardebili
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Pooja Tanty
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| | - Wojciech Wiślicki
- Department of Complex Systems, National Centre for Nuclear Research, 05-400 Otwock-Świerk, Poland
| | - Leszek Królicki
- Department of Nuclear Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Ewa Ł Stępień
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, S. Łojasiewicza 11, 30-348 Krakow, Poland
- Centre for Theranostics, Jagiellonian University, Kopernika 40, 31-501 Krakow, Poland
| |
Collapse
|
44
|
Leuzy A, Raket LL, Villemagne VL, Klein G, Tonietto M, Olafson E, Baker S, Saad ZS, Bullich S, Lopresti B, Bohorquez SS, Boada M, Betthauser TJ, Charil A, Collins EC, Collins JA, Cullen N, Gunn RN, Higuchi M, Hostetler E, Hutchison RM, Iaccarino L, Insel PS, Irizarry MC, Jack CR, Jagust WJ, Johnson KA, Johnson SC, Karten Y, Marquié M, Mathotaarachchi S, Mintun MA, Ossenkoppele R, Pappas I, Petersen RC, Rabinovici GD, Rosa‐Neto P, Schwarz CG, Smith R, Stephens AW, Whittington A, Carrillo MC, Pontecorvo MJ, Haeberlein SB, Dunn B, Kolb HC, Sivakumaran S, Rowe CC, Hansson O, Doré V. Harmonizing tau positron emission tomography in Alzheimer's disease: The CenTauR scale and the joint propagation model. Alzheimers Dement 2024; 20:5833-5848. [PMID: 39041435 PMCID: PMC11497758 DOI: 10.1002/alz.13908] [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: 11/07/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Tau-positron emission tomography (PET) outcome data of patients with Alzheimer's disease (AD) cannot currently be meaningfully compared or combined when different tracers are used due to differences in tracer properties, instrumentation, and methods of analysis. METHODS Using head-to-head data from five cohorts with tau PET radiotracers designed to target tau deposition in AD, we tested a joint propagation model (JPM) to harmonize quantification (units termed "CenTauR" [CTR]). JPM is a statistical model that simultaneously models the relationships between head-to-head and anchor point data. JPM was compared to a linear regression approach analogous to the one used in the amyloid PET Centiloid scale. RESULTS A strong linear relationship was observed between CTR values across brain regions. Using the JPM approach, CTR estimates were similar to, but more accurate than, those derived using the linear regression approach. DISCUSSION Preliminary findings using the JPM support the development and adoption of a universal scale for tau-PET quantification. HIGHLIGHTS Tested a novel joint propagation model (JPM) to harmonize quantification of tau PET. Units of common scale are termed "CenTauRs". Tested a Centiloid-like linear regression approach. Using five cohorts with head-to-head tau PET, JPM outperformed linearregressionbased approach. Strong linear relationship was observed between CenTauRs values across brain regions.
Collapse
|
45
|
Soleimani-Meigooni DN, Smith R, Provost K, Lesman-Segev OH, Allen IE, Chen MK, Cho H, Edwards L, Janelidze S, La Joie R, Mundada N, Ossenkoppele R, Stomrud E, Strandberg O, Strom A, Boxer AL, Dage JL, Gorno-Tempini ML, Kramer JH, Miller BL, Rojas JC, Rosen HJ, Lyoo CH, Hansson O, Rabinovici GD. Head-to-Head Comparison of Tau and Amyloid Positron Emission Tomography Visual Reads for Differential Diagnosis of Neurodegenerative Disorders: An International, Multicenter Study. Ann Neurol 2024; 96:476-487. [PMID: 38888212 PMCID: PMC11324380 DOI: 10.1002/ana.27008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVE We compared the accuracy of amyloid and [18F]Flortaucipir (FTP) tau positron emission tomography (PET) visual reads for distinguishing patients with mild cognitive impairment (MCI) or dementia with fluid biomarker support of Alzheimer's disease (AD). METHODS Participants with FTP-PET, amyloid-PET, and diagnosis of dementia-AD (n = 102), MCI-AD (n = 41), non-AD diseases (n = 76), and controls (n = 20) were included. AD status was determined independent of PET by cerebrospinal fluid or plasma biomarkers. The mean age was 66.9 years, and 44.8% were women. Three readers interpreted scans blindly and independently. Amyloid-PET was classified as positive/negative using tracer-specific criteria. FTP-PET was classified as positive with medial temporal lobe (MTL) binding as the minimum uptake indicating AD tau (tau-MTL+), positive with posterolateral temporal or extratemporal cortical binding in an AD-like pattern (tau-CTX+), or negative. The majority of scan interpretations were used to calculate diagnostic accuracy of visual reads in detecting MCI/dementia with fluid biomarker support for AD (MCI/dementia-AD). RESULTS Sensitivity of amyloid-PET for MCI/dementia-AD was 95.8% (95% confidence interval 91.1-98.4%), which was comparable to tau-CTX+ 92.3% (86.7-96.1%, p = 0.67) and tau-MTL+ 97.2% (93.0-99.2%, p = 0.27). Specificity of amyloid-PET for biomarker-negative healthy and disease controls was 84.4% (75.5-91.0%), which was like tau-CTX+ 88.5% (80.4-94.1%, p = 0.34), and trended toward being higher than tau-MTL+ 75.0% (65.1-83.3%, p = 0.08). Tau-CTX+ had higher specificity than tau-MTL+ (p = 0.0002), but sensitivity was lower (p = 0.02), driven by decreased sensitivity for MCI-AD (80.5% [65.1-91.2] vs. 95.1% [83.5-99.4], p = 0.03). INTERPRETATION Amyloid- and tau-PET visual reads have similar sensitivity/specificity for detecting AD in cognitively impaired patients. Visual tau-PET interpretations requiring cortical binding outside MTL increase specificity, but lower sensitivity for MCI-AD. ANN NEUROL 2024;96:476-487.
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
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Karine Provost
- Department of Nuclear Medicine, Centre Hospitalier de l’Université de Montréal, Montréal, Canada
| | - Orit H. Lesman-Segev
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Isabel Elaine Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Miranda K. Chen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Clinical Psychology, San Diego State University & University of California, San Diego, CA, USA
| | | | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Nidhi Mundada
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Health Sciences and Technology, Harvard & Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam L. Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jeffrey L. Dage
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Joel H. Kramer
- 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
| | - Julio C. Rojas
- 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
| | - Chul H. Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Gil D. Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
46
|
Liu K, Tao Y, Zhao Q, Xia W, Li X, Zhang S, Yao Y, Xiang H, Han C, Tan L, Sun B, Li D, Li A, Liu C. Binding adaptability of chemical ligands to polymorphic α-synuclein amyloid fibrils. Proc Natl Acad Sci U S A 2024; 121:e2321633121. [PMID: 39172784 PMCID: PMC11363296 DOI: 10.1073/pnas.2321633121] [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: 12/08/2023] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
α-synuclein (α-syn) assembles into structurally distinct fibril polymorphs seen in different synucleinopathies, such as Parkinson's disease and multiple system atrophy. Targeting these unique fibril structures using chemical ligands holds diagnostic significance for different disease subtypes. However, the molecular mechanisms governing small molecules interacting with different fibril polymorphs remain unclear. Here, we investigated the interactions of small molecules belonging to four distinct scaffolds, with different α-syn fibril polymorphs. Using cryo-electron microscopy, we determined the structures of these molecules when bound to the fibrils formed by E46K mutant α-syn and compared them to those bound with wild-type α-syn fibrils. Notably, we observed that these ligands exhibit remarkable binding adaptability, as they engage distinct binding sites across different fibril polymorphs. While the molecular scaffold primarily steered the binding locations and geometries on specific sites, the conjugated functional groups further refined this adaptable binding by fine-tuning the geometries and binding sites. Overall, our finding elucidates the adaptability of small molecules binding to different fibril structures, which sheds light on the diagnostic tracer and drug developments tailored to specific pathological fibril polymorphs.
Collapse
Affiliation(s)
- Kaien Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Xiang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Yuxuan Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Huaijiang Xiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Chao Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Li Tan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
| | - Bo Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai201210, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Ang Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai200032, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai201210, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai200032, China
| |
Collapse
|
47
|
Bader I, Groot C, Tan HS, Milongo JMA, Haan JD, Verberk IMW, Yong K, Orellina J, Campbell S, Wilson D, van Harten AC, Kok PHB, van der Flier WM, Pijnenburg YAL, Barkhof F, van de Giessen E, Teunissen CE, Bouwman FH, Ossenkoppele R. Rationale and design of the BeyeOMARKER study: prospective evaluation of blood- and eye-based biomarkers for early detection of Alzheimer's disease pathology in the eye clinic. Alzheimers Res Ther 2024; 16:190. [PMID: 39169442 PMCID: PMC11340081 DOI: 10.1186/s13195-024-01545-1] [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: 05/01/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a common, complex and multifactorial disease that may require screening across multiple routes of referral to enable early detection and subsequent future implementation of tailored interventions. Blood- and eye-based biomarkers show promise as low-cost, scalable and patient-friendly tools for early AD detection given their ability to provide information on AD pathophysiological changes and manifestations in the retina, respectively. Eye clinics provide an intriguing real-world proof-of-concept setting to evaluate the performance of these potential AD screening tools given the intricate connections between the eye and brain, presumed enrichment for AD pathology in the aging population with eye disorders, and the potential for an accelerated diagnostic pathway for under-recognized patient groups. METHODS The BeyeOMARKER study is a prospective, observational, longitudinal cohort study aiming to include individuals visiting an eye-clinic. Inclusion criteria entail being ≥ 50 years old and having no prior dementia diagnosis. Excluded eye-conditions include traumatic insults, superficial inflammation, and conditions in surrounding structures of the eye that are not engaged in vision. The BeyeOMARKER cohort (n = 700) will undergo blood collection to assess plasma p-tau217 levels and a brief cognitive screening at the eye clinic. All participants will subsequently be invited for annual longitudinal follow-up including remotely administered cognitive screening and questionnaires. The BeyeOMARKER + cohort (n = 150), consisting of 100 plasma p-tau217 positive participants and 50 matched negative controls selected from the BeyeOMARKER cohort, will additionally undergo Aβ-PET and tau-PET, MRI, retinal imaging including hyperspectral imaging (primary), widefield imaging, optical coherence tomography (OCT) and OCT-Angiography (secondary), and cognitive and cortical vision assessments. RESULTS We aim to implement the current protocol between April 2024 until March 2027. Primary outcomes include the performance of plasma p-tau217 and hyperspectral retinal imaging to detect AD pathology (using Aβ- and tau-PET visual read as reference standard) and to detect cognitive decline. Initial follow-up is ~ 2 years but may be extended with additional funding. CONCLUSIONS We envision that the BeyeOMARKER study will demonstrate the feasibility of early AD detection based on blood- and eye-based biomarkers in alternative screening settings, and will improve our understanding of the eye-brain connection. TRIAL REGISTRATION The BeyeOMARKER study (Eudamed CIV ID: CIV-NL-23-09-044086; registration date: 19th of March 2024) is approved by the ethical review board of the Amsterdam UMC.
Collapse
Affiliation(s)
- Ilse Bader
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands.
| | - Colin Groot
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - H Stevie Tan
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC Location VUmc, Amsterdam Reproduction and Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Jean-Marie A Milongo
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Jurre den Haan
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Keir Yong
- Queen Square Institute of Neurology, Dementia Research Centre, London, WC1N 3BG, UK
| | | | | | | | - Argonde C van Harten
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Pauline H B Kok
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Wiesje M van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Yolande A L Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Frederik Barkhof
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
- UCL Queen Square Institute of Neurology and Centre for Medical Image Computing, University College, London, WC1N 3BG, UK
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Femke H Bouwman
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Rik Ossenkoppele
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
| |
Collapse
|
48
|
Groot C, Smith R, Collij LE, Mastenbroek SE, Stomrud E, Binette AP, Leuzy A, Palmqvist S, Mattsson-Carlgren N, Strandberg O, Cho H, Lyoo CH, Frisoni GB, Peretti DE, Garibotto V, La Joie R, Soleimani-Meigooni DN, Rabinovici G, Ossenkoppele R, Hansson O. Tau Positron Emission Tomography for Predicting Dementia in Individuals With Mild Cognitive Impairment. JAMA Neurol 2024; 81:845-856. [PMID: 38857029 PMCID: PMC11165418 DOI: 10.1001/jamaneurol.2024.1612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/09/2024] [Indexed: 06/11/2024]
Abstract
Importance An accurate prognosis is especially pertinent in mild cognitive impairment (MCI), when individuals experience considerable uncertainty about future progression. Objective To evaluate the prognostic value of tau positron emission tomography (PET) to predict clinical progression from MCI to dementia. Design, Setting, and Participants This was a multicenter cohort study with external validation and a mean (SD) follow-up of 2.0 (1.1) years. Data were collected from centers in South Korea, Sweden, the US, and Switzerland from June 2014 to January 2024. Participant data were retrospectively collected and inclusion criteria were a baseline clinical diagnosis of MCI; longitudinal clinical follow-up; a Mini-Mental State Examination (MMSE) score greater than 22; and available tau PET, amyloid-β (Aβ) PET, and magnetic resonance imaging (MRI) scan less than 1 year from diagnosis. A total of 448 eligible individuals with MCI were included (331 in the discovery cohort and 117 in the validation cohort). None of these participants were excluded over the course of the study. Exposures Tau PET, Aβ PET, and MRI. Main Outcomes and Measures Positive results on tau PET (temporal meta-region of interest), Aβ PET (global; expressed in the standardized metric Centiloids), and MRI (Alzheimer disease [AD] signature region) was assessed using quantitative thresholds and visual reads. Clinical progression from MCI to all-cause dementia (regardless of suspected etiology) or to AD dementia (AD as suspected etiology) served as the primary outcomes. The primary analyses were receiver operating characteristics. Results In the discovery cohort, the mean (SD) age was 70.9 (8.5) years, 191 (58%) were male, the mean (SD) MMSE score was 27.1 (1.9), and 110 individuals with MCI (33%) converted to dementia (71 to AD dementia). Only the model with tau PET predicted all-cause dementia (area under the receiver operating characteristic curve [AUC], 0.75; 95% CI, 0.70-0.80) better than a base model including age, sex, education, and MMSE score (AUC, 0.71; 95% CI, 0.65-0.77; P = .02), while the models assessing the other neuroimaging markers did not improve prediction. In the validation cohort, tau PET replicated in predicting all-cause dementia. Compared to the base model (AUC, 0.75; 95% CI, 0.69-0.82), prediction of AD dementia in the discovery cohort was significantly improved by including tau PET (AUC, 0.84; 95% CI, 0.79-0.89; P < .001), tau PET visual read (AUC, 0.83; 95% CI, 0.78-0.88; P = .001), and Aβ PET Centiloids (AUC, 0.83; 95% CI, 0.78-0.88; P = .03). In the validation cohort, only the tau PET and the tau PET visual reads replicated in predicting AD dementia. Conclusions and Relevance In this study, tau-PET showed the best performance as a stand-alone marker to predict progression to dementia among individuals with MCI. This suggests that, for prognostic purposes in MCI, a tau PET scan may be the best currently available neuroimaging marker.
Collapse
Affiliation(s)
- Colin Groot
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Lyduine E. Collij
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Sophie E. Mastenbroek
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Giovanni B. Frisoni
- Memory Clinic, Department of Rehabilitation and Geriatrics, Geneva University and University Hospitals, Geneva, Switzerland
- Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
| | - Debora E. Peretti
- Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland
- Center for Biomedical Imaging, Geneva, Switzerland
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, University of California, San Francisco
| | - David N. Soleimani-Meigooni
- Department of Neurology, Memory and Aging Center, University of California, San Francisco
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Gil Rabinovici
- Department of Neurology, Memory and Aging Center, University of California, San Francisco
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
- Associate Editor, JAMA Neurology
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
49
|
Jack CR, Andrews JS, Beach TG, Buracchio T, Dunn B, Graf A, Hansson O, Ho C, Jagust W, McDade E, Molinuevo JL, Okonkwo OC, Pani L, Rafii MS, Scheltens P, Siemers E, Snyder HM, Sperling R, Teunissen CE, Carrillo MC. Revised criteria for diagnosis and staging of Alzheimer's disease: Alzheimer's Association Workgroup. Alzheimers Dement 2024; 20:5143-5169. [PMID: 38934362 PMCID: PMC11350039 DOI: 10.1002/alz.13859] [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: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 06/28/2024]
Abstract
The National Institute on Aging and the Alzheimer's Association convened three separate work groups in 2011 and single work groups in 2012 and 2018 to create recommendations for the diagnosis and characterization of Alzheimer's disease (AD). The present document updates the 2018 research framework in response to several recent developments. Defining diseases biologically, rather than based on syndromic presentation, has long been standard in many areas of medicine (e.g., oncology), and is becoming a unifying concept common to all neurodegenerative diseases, not just AD. The present document is consistent with this principle. Our intent is to present objective criteria for diagnosis and staging AD, incorporating recent advances in biomarkers, to serve as a bridge between research and clinical care. These criteria are not intended to provide step-by-step clinical practice guidelines for clinical workflow or specific treatment protocols, but rather serve as general principles to inform diagnosis and staging of AD that reflect current science. HIGHLIGHTS: We define Alzheimer's disease (AD) to be a biological process that begins with the appearance of AD neuropathologic change (ADNPC) while people are asymptomatic. Progression of the neuropathologic burden leads to the later appearance and progression of clinical symptoms. Early-changing Core 1 biomarkers (amyloid positron emission tomography [PET], approved cerebrospinal fluid biomarkers, and accurate plasma biomarkers [especially phosphorylated tau 217]) map onto either the amyloid beta or AD tauopathy pathway; however, these reflect the presence of ADNPC more generally (i.e., both neuritic plaques and tangles). An abnormal Core 1 biomarker result is sufficient to establish a diagnosis of AD and to inform clinical decision making throughout the disease continuum. Later-changing Core 2 biomarkers (biofluid and tau PET) can provide prognostic information, and when abnormal, will increase confidence that AD is contributing to symptoms. An integrated biological and clinical staging scheme is described that accommodates the fact that common copathologies, cognitive reserve, and resistance may modify relationships between clinical and biological AD stages.
Collapse
Affiliation(s)
| | - J. Scott Andrews
- Global Evidence & OutcomesTakeda Pharmaceuticals Company LimitedCambridgeMassachusettsUSA
| | - Thomas G. Beach
- Civin Laboratory for NeuropathologyBanner Sun Health Research InstituteSun CityArizonaUSA
| | - Teresa Buracchio
- Office of NeuroscienceU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Ana Graf
- NovartisNeuroscience Global Drug DevelopmentBaselSwitzerland
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Faculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University Hospital, MalmöLundSweden
| | - Carole Ho
- DevelopmentDenali TherapeuticsSouth San FranciscoCaliforniaUSA
| | - William Jagust
- School of Public Health and Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Eric McDade
- Department of NeurologyWashington University St. Louis School of MedicineSt. LouisMissouriUSA
| | - Jose Luis Molinuevo
- Department of Global Clinical Development H. Lundbeck A/SExperimental MedicineCopenhagenDenmark
| | - Ozioma C. Okonkwo
- Department of Medicine, Division of Geriatrics and GerontologyUniversity of Wisconsin School of MedicineMadisonWisconsinUSA
| | - Luca Pani
- University of MiamiMiller School of MedicineMiamiFloridaUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research Institute (ATRI)Keck School of Medicine at the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Philip Scheltens
- Amsterdam University Medical Center (Emeritus)NeurologyAmsterdamthe Netherlands
| | - Eric Siemers
- Clinical ResearchAcumen PharmaceuticalsZionsvilleIndianaUSA
| | - Heather M. Snyder
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| | - Reisa Sperling
- Department of Neurology, Brigham and Women's HospitalMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Charlotte E. Teunissen
- Department of Laboratory MedicineAmsterdam UMC, Neurochemistry LaboratoryAmsterdamthe Netherlands
| | - Maria C. Carrillo
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
50
|
Guo T, Li A, Sun P, He Z, Cai Y, Lan G, Liu L, Li J, Yang J, Zhu Y, Zhao R, Chen X, Shi D, Liu Z, Wang Q, Xu L, Zhou L, Ran P, Wang X, Sun K, Lu J, Han Y. Astrocyte reactivity is associated with tau tangle load and cortical thinning in Alzheimer's disease. Mol Neurodegener 2024; 19:58. [PMID: 39080744 PMCID: PMC11290175 DOI: 10.1186/s13024-024-00750-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND It is not fully established whether plasma β-amyloid(Aβ)42/Aβ40 and phosphorylated Tau181 (p-Tau181) can effectively detect Alzheimer's disease (AD) pathophysiology in older Chinese adults and how these biomarkers correlate with astrocyte reactivity, Aβ plaque deposition, tau tangle aggregation, and neurodegeneration. METHODS We recruited 470 older adults and analyzed plasma Aβ42/Aβ40, p-Tau181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) using the Simoa platform. Among them, 301, 195, and 70 underwent magnetic resonance imaging, Aβ and tau positron emission tomography imaging. The plasma Aβ42/Aβ40 and p-Tau181 thresholds were defined as ≤0.0609 and ≥2.418 based on the receiver operating characteristic curve analysis using the Youden index by comparing Aβ-PET negative cognitively unimpaired individuals and Aβ-PET positive cognitively impaired patients. To evaluate the feasibility of using plasma Aβ42/Aβ40 (A) and p-Tau181 (T) to detect AD and understand how astrocyte reactivity affects this process, we compared plasma GFAP, Aβ plaque, tau tangle, plasma NfL, hippocampal volume, and temporal-metaROI cortical thickness between different plasma A/T profiles and explored their relations with each other using general linear models, including age, sex, APOE-ε4, and diagnosis as covariates. RESULTS Plasma A+/T + individuals showed the highest levels of astrocyte reactivity, Aβ plaque, tau tangle, and axonal degeneration, and the lowest hippocampal volume and temporal-metaROI cortical thickness. Lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were independently and synergistically correlated with higher plasma GFAP and Aβ plaque. Elevated plasma p-Tau181 and GFAP concentrations were directly and interactively associated with more tau tangle formation. Regarding neurodegeneration, higher plasma p-Tau181 and GFAP concentrations strongly correlated with more axonal degeneration, as measured by plasma NfL, and lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were related to greater hippocampal atrophy. Higher plasma GFAP levels were associated with thinner cortical thickness and significantly interacted with lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 in predicting more temporal-metaROI cortical thinning. Voxel-wise imaging analysis confirmed these findings. DISCUSSION This study provides a valuable reference for using plasma biomarkers to detect AD in the Chinese community population and offers novel insights into how astrocyte reactivity contributes to AD progression, highlighting the importance of targeting reactive astrogliosis to prevent AD.
Collapse
Affiliation(s)
- Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Anqi Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Pan Sun
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Zhengbo He
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Yue Cai
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Guoyu Lan
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Lin Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jieyin Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jie Yang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yalin Zhu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Ruiyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Dai Shi
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Zhen Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Qingyong Wang
- Department of Neurology, Shenzhen Guangming District People's Hospital, Shenzhen, 518107, China
| | - Linsen Xu
- Department of Medical Imaging, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, China
| | - Liemin Zhou
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Pengcheng Ran
- Department of Nuclear Medicine, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Xinlu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jie Lu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
| | - Ying Han
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China.
- National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
| |
Collapse
|