1
|
Qiang Q, Skudder-Hill L, Toyota T, Huang Z, Wei W, Adachi H. CSF α-synuclein aggregation is associated with APOE ε4 and progressive cognitive decline in Alzheimer's disease. Neurobiol Aging 2025; 150:9-18. [PMID: 40043469 DOI: 10.1016/j.neurobiolaging.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 04/10/2025]
Abstract
At autopsy, around half of the Alzheimer's disease (AD) brains exhibit Lewy body pathology, and the main component of Lewy body pathology is α-synuclein aggregates. This study investigated the prevalence of cerebrospinal fluid (CSF) α-synuclein aggregation and its association with demographic factors and cognitive decline among 1619 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI), with the test for α-synuclein aggregation by seed amplification assay (SAA). This cohort consisted of 595 cognitively normal (CN) individuals, 765 with mild cognitive impairment (MCI), and 259 with AD dementia. The results showed a higher prevalence of positive α-synuclein aggregation status in the AD dementia group (37.07 %) and the MCI group (22.75 %) compared to CN controls (16.13 %). Additionally, APOE ε4 carriers exhibited a higher prevalence of α-synuclein aggregation compared to non-carriers: 20.12 % for APOE ε4-/- (non-carriers), 24.82 % for APOE ε4 + /-, and 30.92 % for APOE ε4 + /+ . Longitudinally, positive CSF α-synuclein aggregation associated with accelerated cognitive decline, especially in the MCI and AD groups. Notably, positive aggregation status did not significantly affect cognitive trajectories in CN individuals. Moreover, APOE ε4 carriers with positive CSF α-synuclein aggregation experienced more pronounced cognitive decline. This study provides evidence that CSF α-synuclein aggregation is associated with cognitive function and the APOE ε4 allele. These findings suggest that CSF α-synuclein SAA, in combination with APOE ε4 status, could serve as biomarkers for predicting cognitive decline in AD.
Collapse
Affiliation(s)
- Qiang Qiang
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, Shanghai, China; Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Loren Skudder-Hill
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tomoko Toyota
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Wenshi Wei
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, Shanghai, China
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan.
| |
Collapse
|
2
|
Zhang Z, Peng J, Shao Y, Li X, Xu Y, Song Q, Xie Y, Shu Z. Use of magnetic resonance structural imaging to identify disease progression in patients with mild cognitive impairment: A voxel-based morphometry and surface-based morphometry study. Neuroscience 2025:S0306-4522(25)00322-7. [PMID: 40318840 DOI: 10.1016/j.neuroscience.2025.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/13/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
Voxel-based morphometry (VBM) and surface-based morphometry (SBM) based on magnetic resonance structural imaging were used to identify disease progression in mild cognitive impairment (MCI) patients. A retrospective analysis was conducted on 154 MCI patients from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, with 62 patients classified into the progressive MCI (pMCI) group and 92 patients into the stable MCI (sMCI) group. VBM and SBM were employed to identify structural differences between sMCI and pMCI patients, and differential features were extracted for model construction. The logistic regression method was used to establish relevant index models, and the DeLong test was used to compare the diagnostic performance of the different models. Additionally, 51 patients from the National Alzheimer's Coordinating Center (NACC) database were used as an external validation set to further validate the clinical efficacy of the model. Significant structural differences between pMCI and sMCI patients were revealed through VBM and SBM analyses. Volume reductions were observed in the frontal and temporal lobes, and cortical thinning occurred in the left inferior and superior parietal cortices. Reduced gyrification was observed in the bilateral insular gyrus. The structural joint model, which combines volume and cortical indices, demonstrated higher diagnostic accuracy compared to the joint scale index model that combines the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MOCA) indices. The findings indicate that combined VBM and SBM analysis offers a sensitive and noninvasive approach to detect structural biomarkers of MCI progression, providing a practical tool for early risk stratification and personalized clinical management.
Collapse
Affiliation(s)
- Zihan Zhang
- Jinzhou Medical University Postgraduate Education Base (Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang Province, China
| | - Jiaxuan Peng
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Yuan Shao
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Xiaotian Li
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Yuyun Xu
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Qiaowei Song
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Yelei Xie
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China
| | - Zhenyu Shu
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
3
|
Rea Reyes RE, Wilson RE, Langhough RE, Studer RL, Jonaitis EM, Oomens JE, Planalp EM, Bendlin BB, Chin NA, Asthana S, Zetterberg H, Johnson SC. Targeted proteomic biomarker profiling using NULISA in a cohort enriched with risk for Alzheimer's disease and related dementias. Alzheimers Dement 2025; 21:e70166. [PMID: 40318118 DOI: 10.1002/alz.70166] [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: 11/27/2024] [Revised: 02/14/2025] [Accepted: 03/11/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION Targeted proteomic assays may be useful for diagnosing and staging Alzheimer's disease and related dementias (ADRD). We evaluated the performance of a 120-marker central nervous system (CNS) NUcleic acid Linked Immuno-Sandwich Assay (NULISA) panel in samples spanning the Alzheimer's disease (AD) spectrum. METHODS Cross-sectional plasma samples (n = 252) were analyzed using NULISAseq CNS panel from Alamar Biosciences. Receiver-operating characteristic (ROC) analyses demonstrated the accuracy from NULISAseq-tau phosphorylated at threonine 217 (pTau217) in detecting amyloid (A) and tau (T) positron emission tomography (PET) positivity. Differentially expressed proteins were identified using volcano plots. RESULTS NULISAseq-pTau217 accurately classified A/T PET status with ROC areas under the curve of 0.92/0.86; pTau217 was upregulated in A+, T+, and impaired groups with log2-fold changes of 1.21, 0.57, and 4.63, respectively, compared to A-. Of interest, TAR DNA-binding protein 43 (TDP-43) phosphorylated at serine 409 (pTDP43-409) was also upregulated in the impaired group and correlated with declining hippocampal volume and cognitive trajectories. DISCUSSION This study shows the potential of a targeted proteomics panel for characterizing brain changes pertinent to ADRD. The promising pTDP43-409 findings require further replication. HIGHLIGHTS The NULISAseq pTau217 assay was comparable to the Simoa pTau217 assay, both utilizing the ALZpath antibody, in detecting amyloid positron emission tomography (PET) positivity, each with areas under the curve greater than 90%. Nineteen proteins were differentially expressed in participants with mild cognitive impairment (MCI) compared to those who were unimpaired. Markers of non-AD proteinopathies such as pTDP43-409, oligomeric alpha-synuclein, and huntingtin (HTT), were among those upregulated in MCI. High levels of plasma pTDP43-409 were associated with worsening hippocampal atrophy and cognitive decline, clinical indicators of limbic-predominant age-related TDP-43 encephalopathy (LATE), compared to those with low pTDP43-409.
Collapse
Affiliation(s)
- Ramiro Eduardo Rea Reyes
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Rachael E Wilson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Rachel L Studer
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Julie E Oomens
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Elizabeth M Planalp
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Nathaniel A Chin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Science Park, Hong Kong, China
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
4
|
Starmans NL, Leeuwis AE, Bennink E, Meyer Viol SL, Golla SS, Dankbaar JW, Bron EE, Biessels GJ, Kappelle LJ, van der Flier WM, Tolboom N. Dynamic PET imaging in patients with unilateral carotid occlusion shows lateralized cerebral hypoperfusion, but no amyloid binding. J Alzheimers Dis 2025:13872877251329593. [PMID: 40241519 DOI: 10.1177/13872877251329593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
BackgroundCarotid occlusive disease is a risk factor for cognitive decline. A possible underlying etiology is that hemodynamic impairment results in decreased cerebral perfusion, exacerbated amyloid-β accumulation (Aβ) and poorer cognitive performance.ObjectiveWe aimed to determine whether patients with unilateral internal carotid artery (ICA) occlusion have less cerebral perfusion and more Aβ in the ipsilateral than in the contralateral hemisphere, and whether perfusion and Aβ are associated with cognitive functioning.MethodsWe included 20 patients (age 67.2 ± 7.0 years, 8 females, MMSE 29 [27-29]) with unilateral ICA occlusion, which underwent neuropsychological assessment and dynamic 18F-Florbetaben positron emission tomography (PET). Global and regional relative perfusion (R1) and binding potential (BPND) were obtained from the PET-images using a simplified reference tissue model. We performed Wilcoxon signed-rank tests to examine differences between hemispheres within subjects and linear regression to investigate associations with cognitive functioning.ResultsMedian global R1 was 0.911 (0.883-0.950) and global BPND was 0.172 (0.129-0.187). R1 was lower in the hemisphere ipsilateral to the ICA occlusion than in the contralateral hemisphere (0.899 [0.876-0.921] versus 0.935 [0.889-0.970]). BPND did not differ significantly between hemispheres (ipsilateral 0.172 [0.124-0.181] versus contralateral 0.168 [0.137-0.191]). Neither cerebral perfusion nor Aβ burden were associated with cognitive functioning.ConclusionsPatients with unilateral ICA occlusion did not have more Aβ in the ipsilateral hemisphere than in the contralateral hemisphere despite ipsilateral hypoperfusion. Perfusion and Aβ were unrelated to cognitive functioning. This indicates that cognitive impairment in patients with ICA occlusion is not due to exacerbated Aβ accumulation.
Collapse
Affiliation(s)
- Naomi Lp Starmans
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna E Leeuwis
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Medical Psychology, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Edwin Bennink
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sebastiaan L Meyer Viol
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Sandeep Sv Golla
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Jan Willem Dankbaar
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Esther E Bron
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L Jaap Kappelle
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
5
|
Vanderlip CR, Taylor L, Kim S, Harris AL, Tuteja N, Meza N, Escalante YY, McMillan L, Yassa MA, Adams JN. Amyloid-Beta Deposition in Basal Frontotemporal Cortex Is Associated with Selective Disruption of Temporal Mnemonic Discrimination. J Neurosci 2025; 45:e1605242025. [PMID: 39843236 PMCID: PMC11884388 DOI: 10.1523/jneurosci.1605-24.2025] [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: 08/23/2024] [Revised: 12/10/2024] [Accepted: 01/03/2025] [Indexed: 01/24/2025] Open
Abstract
Cerebral amyloid-beta (Aβ) accumulation, a hallmark pathology of Alzheimer's disease (AD), precedes clinical impairment by two to three decades. However, it is unclear whether Aβ contributes to subtle memory deficits observed during the preclinical stage. The heterogeneous emergence of Aβ deposition may selectively impact certain memory domains, which rely on distinct underlying neural circuits. In this context, we tested whether specific domains of mnemonic discrimination, a neural computation essential for episodic memory, exhibit specific deficits related to early Aβ deposition. We tested 108 cognitively unimpaired human older adults (66% female) who underwent 18F-florbetapir positron emission tomography (Aβ-PET) and a control group of 35 young adults, on a suite of mnemonic discrimination tasks taxing object, spatial, and temporal domains. We hypothesized that Aβ pathology would be selectively associated with temporal discrimination performance due to Aβ's propensity to accumulate in the basal frontotemporal cortex, which supports temporal processing. Consistent with this hypothesis, we found a dissociation in which generalized age-related deficits were found for object and spatial mnemonic discrimination, while Aβ-PET levels were selectively associated with deficits in temporal mnemonic discrimination. Furthermore, we found that higher Aβ-PET levels in the medial orbitofrontal and inferior temporal cortex, regions supporting temporal processing, were associated with greater temporal mnemonic discrimination deficits, pointing to the selective vulnerability of circuits related to temporal processing early in AD progression. These results suggest that Aβ accumulation within basal frontotemporal regions may disrupt temporal mnemonic discrimination in preclinical AD, and future work is needed to determine whether assessing temporal mnemonic discrimination can aid in predicting emerging AD progression.
Collapse
Affiliation(s)
- Casey R Vanderlip
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Lisa Taylor
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Soyun Kim
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Alyssa L Harris
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Nandita Tuteja
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Novelle Meza
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Yuritza Y Escalante
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Liv McMillan
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Michael A Yassa
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| | - Jenna N Adams
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697
| |
Collapse
|
6
|
Vanderlip CR, Stark CEL. Integrating Plasma pTau-217 and Digital Cognitive Assessments for Early Detection in Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.03.25323297. [PMID: 40093240 PMCID: PMC11908293 DOI: 10.1101/2025.03.03.25323297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Plasma pTau-217 has emerged as a sensitive and specific biomarker for early Alzheimer's disease detection. However, the timeline of pathological changes and the onset of cognitive decline remain unclear. On the other hand, digital cognitive assessments have also shown promise in detecting subtle cognitive changes, but the sensitivity and specificity of these assessments is not fully understood. Here, we investigate whether combining these low-burden tools can improve the identification of cognitively unimpaired individuals at high risk for future cognitive decline. We analyzed 954 amyloid-positive cognitively unimpaired individuals who completed a brief digital cognitive assessment and a blood test for pTau-217, evaluating their ability to identify those at high risk for decline on the Preclinical Alzheimer's Cognitive Composite (PACC) and the Mini-Mental State Exam (MMSE). Further, we investigated whether the predictive value of these measures differed by sex or APOE status. We found that combining memory performance with pTau-217 enhanced the ability to identify individuals who declined on the PACC and MMSE over the next five years, even after controlling for age, sex, education, and baseline cognitive performance. Specifically, individuals with both elevated pTau-217 and low memory performance were at a greater risk for future decline than those with either risk factor alone. Notably, the predictive value of these measures did not differ by sex but was significantly stronger in APOE4 noncarriers compared to carriers. Together, this suggests that combining a brief digital cognitive assessment with plasma pTau-217 provides a reliable and sensitive method for identifying individuals at high risk for future cognitive decline in Alzheimer's disease.
Collapse
Affiliation(s)
- Casey R Vanderlip
- Department of Neurobiology and Behavior, 1424 Biological Sciences III Irvine, University of California Irvine, Irvine, CA, 92697 USA
| | - Craig E L Stark
- Department of Neurobiology and Behavior, 1424 Biological Sciences III Irvine, University of California Irvine, Irvine, CA, 92697 USA
| |
Collapse
|
7
|
Liu S, Maruff P, Saint-Jalmes M, Bourgeat P, Masters CL, Goudey B. Predicting amyloid beta accumulation in cognitively unimpaired older adults: Cognitive assessments provide no additional utility beyond demographic and genetic factors. Alzheimers Dement 2025; 21:e70036. [PMID: 40110649 PMCID: PMC11923568 DOI: 10.1002/alz.70036] [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: 09/20/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Integrating non-invasive measures to estimate abnormal amyloid beta accumulation (Aβ+) is key to developing a screening tool for preclinical Alzheimer's disease (AD). The predictive capability of standard neuropsychological tests in estimating Aβ+ has not been quantified. METHODS We constructed machine learning models using six cognitive measurements alongside demographic and genetic risk factors to predict Aβ status. Data were drawn from three cohorts: Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4), Alzheimer's Disease Neuroimaging Initiative (ADNI), and Australian Imaging, Biomarker & Lifestyle (AIBL) study. Internal validation was conducted within A4 with external validations in ADNI and AIBL to assess model generalizability. RESULTS The highest area under the curve (AUC) for predicting Aβ+ was observed with demographic, genetic, and cognitive variables in A4 (median AUC = 0.745), but this was not significantly different from models without cognitive variables. External validation showed no improvement in ADNI and a slight decrease in AIBL. DISCUSSION Standard neuropsychological tests do not significantly enhance Aβ+ prediction in cognitively unimpaired adults beyond demographic and genetic information. HIGHLIGHTS Standard neuropsychological tests do not significantly improve the prediction of amyloid beta positivity (Aβ+) in cognitively unimpaired older adults beyond demographic and genetic information alone. Across three well-characterized cohorts, machine learning models incorporating cognitive measures failed to significantly improve Aβ+ prediction, indicating the limited relationship between cognitive performance on these tests and the risk of pre-clinical Alzheimer's disease (AD). These findings challenge assumptions about cognitive symptoms preceding Aβ+ screening and emphasize the need for developing more sensitive cognitive tests for early AD detection.
Collapse
Affiliation(s)
- Shu Liu
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- CogState Ltd, Melbourne, Victoria, Australia
| | - Martin Saint-Jalmes
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Pierrick Bourgeat
- Australian eHealth Research Centre, Dutton Park, Queensland, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Benjamin Goudey
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Australia BioCommon, University of Melbourne, North Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Cho SH, Kang H, Ham H, Moon SH, Jang H, Yun J, Lee EH, Shin D, Yim S, Kim BC, Kim HJ, Na DL, Seo SW, Kim JP. Comparison of accumulation rates of beta-amyloid tracers and their relationship with cognitive changes. Sci Rep 2025; 15:7072. [PMID: 40016250 PMCID: PMC11868567 DOI: 10.1038/s41598-025-90642-8] [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: 10/05/2024] [Accepted: 02/14/2025] [Indexed: 03/01/2025] Open
Abstract
We aimed to compare amyloid-β (Aβ) accumulation rates between different tracers and investigate whether the relationship between changes in Aβ uptake and cognitive decline varies depending on tracer type. Two cohorts were analyzed: (1) a head-to-head longitudinal cohort using 18F-Florbetaben (FBB) and 18F-Flutemetamol (FMM) tracers (n = 13), and (2) separate longitudinal cohorts for each tracer (n = 174 for both FMM and FBB), matched by propensity score. Aβ uptake was measured using regional direct comparison of Centiloid (rdcCL) values. In the head-to-head cohort, subtracting changes in FMM rdcCL from FBB rdcCL yielded median values above zero in all regions except the cingulate. In the individual tracer cohorts, FBB rdcCL showed faster accumulation than FMM rdcCL in all cortical regions except the striatum (β [SE] = - 2.49 to - 1.56 [0.47-0.54], p < 0.001). Mini-Mental State Examination changes were associated with annualized FMM rdcCL changes in the temporal cortex (p = 0.02) and striatum (p = 0.01); however, no such differences were found in the FBB cohort. Our findings suggest that longitudinal Aβ positron emission tomography studies should consider the specific characteristics of tracers depending on the context of use.
Collapse
Grants
- RS-2022-KH127756 Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare and Ministry of science and ICT, Republic of Korea
- RS-2022-KH127756 Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare and Ministry of science and ICT, Republic of Korea
- RS-2022-KH127756 Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare and Ministry of science and ICT, Republic of Korea
- RS-2022-KH127756 Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare and Ministry of science and ICT, Republic of Korea
- 2024-ER1003-00 the "Korea National Institute of Health" research project
- 2024-ER1003-00 the "Korea National Institute of Health" research project
- NRF-2019R1A5A2027340 the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT)
- NRF-2019R1A5A2027340 the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT)
- No.RS-2021-II212068 Institute of Information & communications Technology Planning & Evaluation (IITP) grant funded by the Korea government (MSIT)
- #SMX1240561 Artificial Intelligence Innovation Hub); Future Medicine 20*30 Project of the Samsung Medical Center
- Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare and Ministry of science and ICT, Republic of Korea
- the “Korea National Institute of Health” research project
- Institute of Information & communications Technology Planning & Evaluation (IITP) grant funded by the Korea government (MSIT)
Collapse
Affiliation(s)
- Soo Hyun Cho
- Department of Neurology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Heekyoung Kang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Hongki Ham
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyemin Jang
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jihwan Yun
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon-si, Republic of Korea
| | - Eun Hye Lee
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Daeun Shin
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sohyun Yim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Byeong Chae Kim
- Department of Neurology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hee Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Alzheimer Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Science and Technology, Samsung Advanced Institute for Health Science & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Duk L Na
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Happymind Clinic, Seoul, Republic of Korea
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
- Samsung Alzheimer Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Health Science and Technology, Samsung Advanced Institute for Health Science & Technology, Sungkyunkwan University, Seoul, Republic of Korea.
| | - Jun Pyo Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
- Samsung Alzheimer Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Cai Y, Fang L, Li A, Yang J, Zhou X, He Z, Sun P, Wang Q, Guo T. Educational attainment, Aβ, tau, and structural brain reserve in Alzheimer's disease. Alzheimers Dement 2025; 21:e14400. [PMID: 39854134 PMCID: PMC11848334 DOI: 10.1002/alz.14400] [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: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 01/26/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) patients with higher educational attainment (EA) often exhibit better cognitive function. However, the relationship among EA status, AD pathology, structural brain reserve, and cognitive decline requires further investigation. METHODS We compared cognitive performance across different amyloid beta (Aβ) positron emission tomography (A ±) statuses and EA levels (High EA/Low EA). We examined the effects of Aβ plaques, tau tangles, and gray matter volume (GMV) on the relationship between EA and domain-specific cognitive decline. RESULTS A+/High-EA individuals exhibited slower cognitive decline in global cognition and language domains than A+/Low-EA individuals. This cognitive benefit was independently and synergistically explained by reduced AD pathology, including lower Aβ and tau burdens, as well as preserved GMV. Additionally, High-EA individuals experienced a median delay of 1.9 years in the onset of significant brain atrophy among A+ individuals. DISCUSSION These findings highlight the independent and synergistic contributions of EA-associated AD pathology and GMV alterations to longitudinal cognitive decline. HIGHLIGHTS Alzheimer's disease (AD) individuals with high educational attainment (EA) show slower declines in global cognition and language. EA-related slower cognitive decline is linked to reduced tau and greater gray matter volume in AD. AD individuals with high EA show a median 1.9 year delayed onset of brain atrophy.
Collapse
Affiliation(s)
- Yue Cai
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
| | - Lili Fang
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
| | - Anqi Li
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
- Division of Life ScienceThe Hong Kong University of Science and TechnologyHKSARChina
| | - Jie Yang
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Xin Zhou
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
- School of Biomedical EngineeringHainan UniversityHaikouChina
| | - Zhengbo He
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Pan Sun
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
| | - Qingyong Wang
- Department of NeurologyShenzhen Guangming District People's HospitalShenzhenChina
| | - Tengfei Guo
- Institute of Neurological and Psychiatric DisordersShenzhen Bay LaboratoryShenzhenChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- Institute of Biomedical EngineeringPeking University Shenzhen Graduate SchoolShenzhenChina
| | | |
Collapse
|
10
|
Rea Reyes RE, Cody KA, Wilson RE, Zetterberg H, Chin NA, Jonaitis EM, Bahr M, Mandel O, Wintlend M, Bendlin BB, Okonkwo OC, Clark LR, Zammit M, Asthana S, Christian BT, Betthauser TJ, Eisenmenger L, Langhough RE, Johnson SC. Visual read of [F-18]florquinitau PET that includes and extends beyond the mesial temporal lobe is associated with increased plasma pTau217 and cognitive decline in a cohort that is enriched with risk for Alzheimer's disease. Alzheimers Dement 2025; 21:e14406. [PMID: 39560002 PMCID: PMC11848396 DOI: 10.1002/alz.14406] [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: 08/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION Patterns of signal from tau positron emission tomography (tau-PET) confined to the medial temporal lobe (MTL) or extended into the neocortex may be relevant for Alzheimer's disease (AD) research if they are linked to differential biomarker levels and cognitive decline. METHODS Visual assessment of Tau-PET [F-18]florquinitau (FQT) exams from 728 initially non-demented older adults yielded four uptake groups: tau-negative (T-), MTL-only (T+MTL), neocortex-only (T+Neo), or both (T+MTL&Neo). Mixed effects models assessed group differences in retrospective cognitive and plasma pTau217 trajectories. RESULTS T+MTL&Neo was the most common T+ group (n = 97; 93% A+) and exhibited the sharpest worsening in cognitive and pTau217 trajectories before tau PET. DISCUSSION The T+MTL&Neo category represents an intermediate to advanced stage of AD preceded by rising ptau217 and progressive cognitive decline. The pTau217 finding suggests that A+, T+ in MTL or neocortex could represent early AD stages, with a higher likelihood of progressing to more advanced stages. HIGHLIGHTS Visual assessments of Tau-PET FQT revealed four distinct uptake groups: tau-negative (T-), MTL-only (T+MTL), neocortex-only (T+Neo), or both (T+MTL&Neo). Amyloid positive participants in the T+MTL and T+MTL&Neo categories showed a retrospectively faster decline in their cognitive trajectories, and a sharper increase in pTau217 levels in plasma, compared to T-. The T+MTL&Neo group displayed sharper trajectories compared with the other Tau positive groups in both their cognitive scores and pTau217 plasma levels. Our results suggest that participants with Tau present in both MTL and neocortex represent an intermediate to advanced stage of AD, whereas participants with signals confined to either MTL or neocortex could represent earlier AD stages.
Collapse
Affiliation(s)
- Ramiro Eduardo Rea Reyes
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Karly A. Cody
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Neurology and Neurological SciencesStanford UniversityPalo AltoCaliforniaUSA
| | - Rachael E. Wilson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water Bay, Science ParkHong KongChina
| | - Nathaniel A. Chin
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Erin M. Jonaitis
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Melissa Bahr
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Olivia Mandel
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Madilynn Wintlend
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Barbara B. Bendlin
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Lindsay R. Clark
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton Memorial Veterans HospitalMadisonWisconsinUSA
| | - Matt Zammit
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bradley T. Christian
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Tobey J. Betthauser
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Laura Eisenmenger
- Department of RadiologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Rebecca E. Langhough
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| |
Collapse
|
11
|
Huang Q, Bolt D, Jonaitis E, Hermann B, Studer R, Du L, Ryther B, Sparks L, Galvin JE, Johnson S, Langhough R. Performance of study partner reports in a non-demented at-risk sample. Alzheimers Dement 2025; 21:e14470. [PMID: 39711292 PMCID: PMC11851318 DOI: 10.1002/alz.14470] [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: 08/14/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION The Clinical Dementia Rating (CDR) Scale is a gold standard for staging impairment in Alzheimer's disease and other dementias (ADRD). The Quick Dementia Rating System (QDRS) offers similar results in 3 to 5 minutes without a trained clinician. This study aimed to (1) investigate concordance between comparably derived QDRS and CDR global scores, (2) examine item-level QDRS/CDR agreement, and (3) compare sample characteristics and cognitive performance across QDRS/CDR global concordant/discordant groups. METHODS The study included 351 QDRS/CDR pairs from 297 participants in the Wisconsin Registry for Alzheimer's Prevention (WRAP). Analyses included descriptive indices of QDRS/CDR agreement, lasso logistic regression, tetrachoric correlations, and linear mixed models. RESULTS The QDRS global/CDR global concordance rate is 70.66%. Memory item discrepancies were primarily responsible for QDRS/CDR global rating discordance. Average cognitive scores were highest in concordant-normal QDRS/CDR and lowest in concordant-abnormal QDRS/CDR. DISCUSSION The QDRS effectively screened for impairment in this sample. Future analyses will investigate QDRS relations to ADRD biomarkers. HIGHLIGHTS The Quick Dementia Rating System (QDRS) effectively screened for impairment in Alzheimer's disease and other dementias (ADRD) in a non-demented sample. Concordance rate between QDRSCDR global and Clinical Dementia Rating (CDR) Scale global scores is 70.66%. Memory item discrepancies primarily cause QDRS/CDR global score discordance. Cognitive scores are associated with QDRS/CDR concordances/discordances. Future analyses will explore QDRS relations to ADRD biomarkers.
Collapse
Affiliation(s)
- Qi Huang
- Department of Educational PsychologyUniversity of Wisconsin–Madison School of EducationMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - Daniel Bolt
- Department of Educational PsychologyUniversity of Wisconsin–Madison School of EducationMadisonWisconsinUSA
| | - Erin Jonaitis
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bruce Hermann
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of NeurologySchool of Medicine and Public Health, University of Wisconsin–MadisonMadisonWisconsinUSA
| | - Rachel Studer
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - Lianlian Du
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush Medical CollegeChicagoIllinoisUSA
| | - Brenda Ryther
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - Lia Sparks
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - James E. Galvin
- Comprehensive Center for Brain HealthDepartment of NeurologyUniversity of Miami, Miller School of MedicineBoca RatonFloridaUSA
| | - Sterling Johnson
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Geriatrics Research, Education and Clinical Center (GRECC)William S. Middleton Memorial Veterans HospitalMadisonWisconsinUSA
| | - Rebecca Langhough
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin–Madison School of Medicine and Public HealthMadisonWisconsinUSA
| |
Collapse
|
12
|
Trelle AN, Young CB, Vossler H, Ramos Benitez J, Cody KA, Mendiola JH, Swarovski MS, Guen YL, Feinstein I, Butler RR, Channappa D, Romero A, Park J, Shahid‐Besanti M, Corso NK, Chau K, Smith AN, Skylar‐Scott I, Yutsis MV, Fredericks CA, Tian L, Younes K, Kerchner GA, Deutsch GK, Davidzon GA, Sha SJ, Henderson VW, Longo FM, Greicius MD, Wyss‐Coray T, Andreasson KI, Poston KL, Wagner AD, Mormino EC, Wilson EN. Plasma Aβ 42/Aβ 40 is sensitive to early cerebral amyloid accumulation and predicts risk of cognitive decline across the Alzheimer's disease spectrum. Alzheimers Dement 2025; 21:e14442. [PMID: 39713875 PMCID: PMC11848181 DOI: 10.1002/alz.14442] [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: 08/06/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION The availability of amyloid beta (Aβ) targeting therapies for Alzheimer's disease (AD) is increasing the demand for scalable biomarkers that are sensitive to early cerebral Aβ accumulation. METHODS We evaluated fully-automated Lumipulse plasma Aβ42/Aβ40 immunoassays for detecting cerebral Aβ in 457 clinically unimpaired (CU) and clinically impaired (CI) Stanford Alzheimer's Disease Research Center (Stanford ADRC) participants and 186 CU in the Stanford Aging and Memory Study (SAMS). Longitudinal change in ADRC plasma Aβ42/Aβ40 and cognition and cross-sectional associations with SAMS memory and tau positron emission tomography (PET) were examined. RESULTS Plasma Aβ42/Aβ40 exhibited high performance in detecting amyloid positivity defined by PET (area under the curve [AUC]: 0.885, 95% confidence interval [CI]: 0.816-0.955). Once abnomal, plasma Aβ42/Aβ40 remained low and predicted cognitive decline in both CU and CI individuals. Among SAMS CU, plasma Aβ42/Aβ40 was associated with poorer hippocampal-dependent memory and elevated tau accumulation. DISCUSSION Lumipulse plasma Aβ42/Aβ40 is a scalable assay for detection of cerebral Aβ and prediction of risk for cognitive decline across the AD continuum. HIGHLIGHTS Lumipulse plasma amyloid beta (Aβ)42/Aβ40 exhibited high accuracy in detecting amyloid positivity. Plasma amyloid-positive (Aβ+) individuals exhibited stability of Aβ42/Aβ40 over time. Plasma Aβ42/Aβ40 predicted future cognitive decline across the Alzheimer's disease (AD) spectrum. Plasma Aβ42/Aβ40 was sensitive to memory and tau burden in clinically unimpaired older adults.
Collapse
|
13
|
Pan F, Huang Q, Huang C, Lu Y, Cui L, Huang L, Guan Y, Xie F, Guo Q. Associations of hippocampal volumes, brain hypometabolism, and plasma NfL with amyloid, tau, and cognitive decline. Alzheimers Dement 2025; 21:e70005. [PMID: 39989286 PMCID: PMC11848211 DOI: 10.1002/alz.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Various indicators of neurodegeneration (N) are used in the assessment of neuronal injury in Alzheimer's disease (AD). The heterogeneity of such indicators is less clear. METHODS A total of 416 individuals with different cognitive statuses were recruited for this study. Differential associations of hippocampal volume (HV), 18F-fluorodeoxyglucose positron emission tomography (FDG PET) standardized uptake value ratios (SUVRs), and plasma neurofilament light chain (NfL) levels with amyloid beta (Aβ)-tau pathology and cognitive impairment were examined. RESULTS HV decreased early during the high Aβ burden but tau-negative stage. FDG PET SUVRs and plasma NfL levels notably changed at tau-positive stages. HV and plasma NfL correlated with cognitive scores in the early to middle stages, while FDG PET SUVRs aligned with cognitive decline from the middle to late stages. Hippocampal atrophy and inferior parietal hypometabolism increased the risk of cognitive impairment in A+T+, while adding NfL+ had no additional impact within the distinct A/T groups. DISCUSSION Different indicators of N have varying relationships to AD pathology and cognitive impairment. HIGHLIGHTS Hippocampal atrophy emerges early with a high amyloid beta burden and exacerbates during the tau-positive phase. Brain hypometabolism and elevated plasma neurofilament light chain (NfL) levels appear mainly in tau-positive stages. Hippocampal volume and plasma NfL levels correlate with cognitive decline in the early to middle stages, while 18F-fluorodeoxyglucose positron emission tomography standardized uptake value ratios in the middle to late stages. Hippocampal atrophy and inferior parietal hypometabolism raise the risk of cognitive impairment in amyloid/tau-positive individuals while adding NfL+ shows no additional effect.
Collapse
Affiliation(s)
- Feng‐Feng Pan
- Department of GerontologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qi Huang
- PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Chu‐Chung Huang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education)Affiliated Mental Health Center (ECNU)School of Psychology and Cognitive ScienceEast China Normal UniversityShanghaiChina
| | - Yao Lu
- Department of GerontologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liang Cui
- Department of GerontologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lin Huang
- Department of GerontologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yihui Guan
- PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Fang Xie
- PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Qi‐Hao Guo
- Department of GerontologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
14
|
Sheng ZH, Liu JY, Ma JY, Mi YC, Wang H, Guo F, Ma LZ, Tan L. Frailty increases the risk of Alzheimer's disease in non-demented individuals: A longitudinal cohort study. J Alzheimers Dis 2025; 103:1023-1035. [PMID: 39956938 DOI: 10.1177/13872877241309081] [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: 02/18/2025]
Abstract
BACKGROUND Frailty, which is considered a potential modifiable risk factor for dementia, continues to generate debate when it comes to Alzheimer's disease (AD). Furthermore, the underlying pathological mechanisms linking frailty to AD remain uncertain. OBJECTIVE We aimed to investigate the relationship between frailty and the risk of AD while elucidating the connections between frailty, AD biomarkers, and cognitive function. METHODS Total of 829 non-frail (261 robust, 568 pre-frail) and 94 frail individuals from the Alzheimer's Disease Neuroimaging Initiative database were recruited. Kaplan-Meier analysis and Cox regression assessed AD risk across diverse frail statuses in 923 non-demented individuals. Multiple linear regression, mixed effects models and causal mediation analyses bootstrapped 10,000 iterations were conducted to examined underlying associations. RESULTS The frail group had a 67.7% increased risk of AD than non-frail group (HR = 1.677; 95%CI, 1.179-2.385; p = 0.004), a 61.8% increased risk of AD than pre-frail group (HR = 1.618; 95%CI, 1.131-2.316; p = 0.009) and a far higher risk of AD than robust group (HR = 2.011; 95%CI, 1.263-3.202; p = 0.003). Frailty was associated with cognitive decline (global cognition, memory and executive function), whole brain and hippocampus atrophy, and ventricle dilation. Higher frail degree predicted faster cognitive decline, brain atrophy and ventricle dilation. Frailty's association with cognition was partially mediated by volume of whole brain (29.54%-30.17% of total effect), hippocampus (18.21%-24.55% of total effect), and ventricle (baseline, 7.62%-10.87% of total effect; change rate, 13.30%-24.33% of total effect). CONCLUSIONS Frailty as a potential risk factor for AD, further mechanisms investigation is warranted; mitigating frailty could potentially contribute to AD prevention.
Collapse
Affiliation(s)
- Ze-Hu Sheng
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jia-Yao Liu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jun-Yi Ma
- Shandong First Medical University, Jinan, China
| | - Yin-Chu Mi
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Hao Wang
- Department of Neurology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Fan Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Jiang Y, Li W, Ma Y, Hou Y. Neuroinflammation-targeted magnetic resonance imaging nanoprobes for the early diagnosis of Alzheimer's disease. J Mater Chem B 2025; 13:1424-1436. [PMID: 39686760 DOI: 10.1039/d4tb02210f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Alzheimer's disease (AD) that is an important contributor to dementia, is a chronic and irreversible neurodegenerative disease, with high rates of disability and mortality. Recently, more and more therapeutic methods have been developed to delay the progression of AD, but it remains a great challenge to achieve the early diagnosis of AD. In this work, we developed a magnetic resonance imaging (MRI) nanoprobe (NP@angiopep-2/CD137) based on angiopep-2 peptide and CD137 antibody with a NaGdF4 nanoparticle as the core and realized neuroinflammation-targeted imaging on APP/PS1 model mice using a clinical 7.0 T MRI scanner. CD137 expression was upregulated in neuroglial cells and cerebral vascular endothelial cells in inflammatory state. In the APP/PS1 mouse model, after administration, the nanoprobe-enhanced images showed specific dot-like signals in the susceptibility-weighted imaging (SWI) sequence. In summary, we designed and synthesized NP@angiopep-2/CD137 nanoprobes using the activation-dependent expression of CD137, which were applied to the pathological assessment of AD based on the hypothesis of AD neuroinflammation, and provided a reliable idea for the early molecular imaging diagnosis of AD.
Collapse
Affiliation(s)
- Yanjiao Jiang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Wenyue Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Yuqiang Ma
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| |
Collapse
|
16
|
Ao J, Picard C, Auld D, Zetterberg H, Brinkmalm A, Blennow K, Villeneuve S, Breitner JCS, Poirier J. Novel synaptic markers predict early tau pathology and cognitive deficit in an asymptomatic population at risk of Alzheimer's disease. Mol Psychiatry 2025:10.1038/s41380-024-02884-z. [PMID: 39827219 DOI: 10.1038/s41380-024-02884-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Cognitive dysfunction in Alzheimer's disease (AD) correlates closely with pathology in the neuronal microtubule-associated protein tau. Tau pathology may spread via neural synapses. In a population of cognitively unimpaired elderly at elevated risk of AD, we investigated four cerebrospinal (CSF) markers of synaptic dysfunction and degeneration. Three of these (SYT1, SNAP25, and ADAM23) are derived from pre-synaptic structures, while ADAM22 reflects post-synaptic changes. All four markers correlated strongly with tau protein measures. In statistical models, SYT1 accounted for more than half the total variance in both total- and P(181)-tau levels. Observed correlations with CSF levels of Alzheimer amyloid-β (Aβ42) were somewhat weaker. In longitudinal data, baseline levels of ADAM22 and ADAM23 robustly predicted increase over time in both total- and P-tau. CSF SYT1 levels also correlated with PET image uptake of tau and (at a trend level) Aβ in areas of interest for early AD pathology. CSF SYT1 and SNAP25 levels correlated inversely with a global psychometric score and several of its domain subscales. In quantitative trait loci analyses, all four synaptic markers were associated with at least one AD genetic risk locus. Upon "staging" participants by their evidence of amyloid and tau pathology (A/T/N framework), the CSF synaptic markers were unexpectedly reduced in participants with CSF evidence of amyloid but not tau pathology. They were clearly elevated, however, in the CSF of persons with indications of both tau and amyloid pathology. These observations provide evidence for clear pre-synaptic degeneration in cognitively unimpaired persons with biomarker evidence of early AD pathology.
Collapse
Affiliation(s)
- Jiarui Ao
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Daniel Auld
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ann Brinkmalm
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - John C S Breitner
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, QC, Canada.
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada.
| |
Collapse
|
17
|
Stephenson HG, Betthauser TJ, Langhough R, Jonaitis E, Du L, Van Hulle C, Kollmorgen G, Quijano‐Rubio C, Chin NA, Okonkwo OC, Carlsson CM, Asthana S, Johnson SC, Blennow K, Zetterberg H, Bendlin BB. Amyloid is associated with accelerated atrophy in cognitively unimpaired individuals. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70089. [PMID: 39996035 PMCID: PMC11848556 DOI: 10.1002/dad2.70089] [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: 08/16/2024] [Revised: 12/13/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION This study examined the association of longitudinal atrophy with baseline cerebrospinal fluid (CSF) amyloid beta (Aβ, A) and phosphorylated tau (p-tau, T) biomarkers (Aβ42/40, p-tau181) in 406 cognitively unimpaired (CU) individuals (6.670 years of follow-up on average, up to 13 imaging visits) to assess whether A+ is associated with Alzheimer's disease-like atrophy and whether this depends on p-tau181 levels. METHODS An A-T- CU group free from abnormal neurodegeneration (N) was identified using a robust normative approach and used to model normal age-related atrophy via z-scoring. Linear mixed-effects models tested differences in longitudinal atrophy between A+ and A-T-N- individuals and between A/T subgroups. RESULTS A+ was associated with worse atrophy within and beyond the medial temporal lobe, even at low levels of p-tau181. DISCUSSION Neurodegeneration likely begins soon after the onset of abnormal Aβ pathology. Clinical intervention at the earliest signs of Aβ pathology may be needed to mitigate further neurodegeneration. Highlights An A-T-N- control group was identified using a robust normative approachA+ was associated with accelerated atrophy in cognitively unimpaired individualsAtrophy was observed even at low p-tau181 levels.
Collapse
|
18
|
Aye S, Johansson G, Hock C, Lannfelt L, Sims JR, Blennow K, Frederiksen KS, Graff C, Molinuevo JL, Scheltens P, Palmqvist S, Schöll M, Wimo A, Kivipelto M, Handels R, Frölich L, Zilka N, Tolar M, Johannsen P, Jönsson L, Winblad B. Point of view: Challenges in implementation of new immunotherapies for Alzheimer's disease. J Prev Alzheimers Dis 2025; 12:100022. [PMID: 39800469 DOI: 10.1016/j.tjpad.2024.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 05/02/2025]
Abstract
The advancement of disease-modifying treatments (DMTs) for Alzheimer's disease (AD), along with the approval of three amyloid-targeting therapies in the US and several other countries, represents a significant development in the treatment landscape, offering new hope for addressing this once untreatable chronic progressive disease. However, significant challenges persist that could impede the successful integration of this class of drugs into clinical practice. These challenges include determining patient eligibility, appropriate use of diagnostic tools and genetic testing in patient care pathways, effective detection and monitoring of side effects, and improving the healthcare system's readiness by engaging both primary care and dementia specialists. Additionally, there are logistical concerns related to infrastructure, as well as cost-effectiveness and reimbursement issues. This article brings together insights from a diverse group of international researchers and dementia experts and outlines the potential challenges and opportunities, urging all stakeholders to prepare for the introduction of DMTs. We emphasize the need to develop appropriate use criteria, including patient characteristics, specifically for the European healthcare system, to ensure that treatments are administered to the most suitable patients. It is crucial to improve the skills and knowledge of physicians to accurately interpret biomarker results, share decision-making with patients, recognize treatment-related side effects, and monitor long-term treatment. We advocate for investment in patient registries and unbiased follow-up studies to better understand treatment effectiveness, evaluate treatment-related side effects, and optimize long-term treatment. Utilizing amyloid-targeting therapies as a starting point for combination therapies should also be a priority.
Collapse
Affiliation(s)
- Sandar Aye
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64 Solna, Sweden.
| | - Gunilla Johansson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64 Solna, Sweden
| | | | - Lars Lannfelt
- Dept. of Public Health, Geriatrics, Uppsala University, Sweden; BioArctic AB, Stockholm, Sweden
| | - John R Sims
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Kaj Blennow
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Kristian S Frederiksen
- Danish Dementia Research Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Graff
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64 Solna, Sweden; Theme Inflammation and Aging, Unit for hereditary dementias Karolinska University Hospital Solna, Sweden
| | - José Luis Molinuevo
- Global Clinical Development, H. Lundbeck A/S, 2500 Valby, Denmark; BarcelonaBeta Brain Research Center, 08005 Barcelona, Spain
| | | | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Psychiatry, Cognition and Aging Psychiatry, Sahlgrenska University Hospital, Mölndal, Sweden; Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - Anders Wimo
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64 Solna, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Finland; The Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, United Kingdom
| | - Ron Handels
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64 Solna, Sweden; Department of Psychiatry and Neuropsychology, Maastricht University, Alzheimer Centre Limburg, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, 6200 MD, Maastricht, the Netherlands
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Norbert Zilka
- Axon Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02 Bratislava, Slovakia
| | - Martin Tolar
- Alzheon, Inc., 111 Speen Street, Framingham, MA, USA
| | - Peter Johannsen
- Medical & Science, Clinical Drug Development. Novo Nordisk A/S, DK-2860 Soeborg, Denmark
| | - Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64 Solna, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64 Solna, Sweden; Theme Inflammation and Aging, Karolinska University Hospital, 141 86 Stockholm, Sweden
| |
Collapse
|
19
|
Huijbers W, Pinter NK, Spaltman M, Cornelis M, Schmand B, Alnaji B, Yargeau M, Harlock S, Dorn RP, Ajtai B, Westphal ES, van Elswijk G. Clinical validity of IntelliSpace Cognition digital assessment platform in mild cognitive impairment. Front Psychol 2024; 15:1451843. [PMID: 39807355 PMCID: PMC11726315 DOI: 10.3389/fpsyg.2024.1451843] [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: 06/19/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
We evaluated a digital cognitive assessment platform, Philips IntelliSpace Cognition, in a case-control study of patients diagnosed with mild cognitive impairment (MCI) and cognitively normal (CN) older adults. Performance on individual neuropsychological tests, cognitive z-scores, and Alzheimer's disease (AD)-specific composite scores was compared between the CN and MCI groups. These groups were matched for age, sex, and education. Performance on all but two neuropsychological tests was worse in the MCI group. After ranking the cognitive scores by effect size, we found that the memory score was the most impaired, followed by executive functioning. The Early AD/MCI Alzheimer's Cognitive Composite (EMACC) and Preclinical Alzheimer's Cognitive Composite (PACC) scores were constructed from the digital tests on Philips IntelliSpace Cognition. Both AD-specific composite scores showed greater sensitivity and specificity than the Mini-Mental State Examination or individual cognitive z-scores. Together, these results demonstrate the diagnostic value of Philips IntelliSpace Cognition in patients with MCI.
Collapse
Affiliation(s)
| | - Nandor K. Pinter
- Dent Neurologic Institute, Amherst, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | | | - Mike Cornelis
- Digital Cognitive Dx, Philips, Eindhoven, Netherlands
| | - Ben Schmand
- Faculty of Social and Behavioral Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Baraa Alnaji
- Dent Neurologic Institute, Amherst, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | | | - Sarah Harlock
- Dent Neurologic Institute, Amherst, NY, United States
| | - Ryu Platinum Dorn
- Dent Neurologic Institute, Amherst, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Bela Ajtai
- Dent Neurologic Institute, Amherst, NY, United States
| | | | | |
Collapse
|
20
|
Jang H, Chun MY, Yun J, Kim JP, Kang SH, Kim HJ, Na DL, Lee EH, Shin D, Ham H, Gu Y, Kim CH, Woo SY, Seo SW. Distinct Cognitive Trajectories According to Amyloid Positivity in Non-Alzheimer Disease Dementias. Clin Nucl Med 2024; 49:1073-1078. [PMID: 39385364 DOI: 10.1097/rlu.0000000000005457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
BACKGROUND The clinical effects of β-amyloid positivity (Aβ+) on copathologies in various dementias remain relatively underexamined. Thus, the present study was conducted to investigate the prevalence and clinical effects of Aβ+ in subcortical vascular cognitive impairment (SVCI) and frontotemporal dementia (FTD). PATIENTS AND METHODS We enrolled SVCI (n = 583), FTD (n = 152), and cognitively unimpaired (CU) participants (n = 1,249) who underwent Aβ PET scans. The odds of having Aβ+ were subsequently compared among the diagnostic groups (CU, SVCI, and FTD) according to age and apolipoprotein E genotype. Additionally, a linear mixed-effects model was used to investigate the effects of Aβ+ on cognitive trajectories in SVCI and FTD. RESULTS Compared with CU, the SVCI group had a higher prevalence of Aβ+ in the 75 to 90 years age group (adjusted odds ratio, 1.97; 95% confidence interval, 1.36-2.85; P < 0.001), as well as within the apolipoprotein E ε3/ε3 group (adjusted odds ratio, 1.78; 95% confidence interval, 1.20-2.63; P = 0.001), whereas the FTD group showed no difference in Aβ+ prevalence. Aβ+ was associated with a worse cognitive trajectory in SVCI (adjusted β-coefficient = -0.6424; P < 0.001), but not in FTD. CONCLUSIONS These findings contribute to our understanding of Aβ biomarker traits in various dementias in Korea.
Collapse
Affiliation(s)
| | | | | | - Jun Pyo Kim
- From the Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | | | | | - Eun Hye Lee
- From the Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Daeun Shin
- From the Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | - Chi-Hun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Sook-Young Woo
- Biomedical Statistics Center, Data Science Research Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | | |
Collapse
|
21
|
An C, Cai H, Ren Z, Fu X, Quan S, Jia L. Biofluid biomarkers for Alzheimer's disease: past, present, and future. MEDICAL REVIEW (2021) 2024; 4:467-491. [PMID: 39664082 PMCID: PMC11629312 DOI: 10.1515/mr-2023-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/04/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD) is a gradually progressive neurodegenerative disease with tremendous social and economic burden. Therefore, early and accurate diagnosis is imperative for effective treatment or prevention of the disease. Cerebrospinal fluid and blood biomarkers emerge as favorable diagnostic tools due to their relative accessibility and potential for widespread clinical use. This review focuses on the AT(N) biomarker system, which includes biomarkers reflecting AD core pathologies, amyloid deposition, and pathological tau, as well as neurodegeneration. Novel biomarkers associated with inflammation/immunity, synaptic dysfunction, vascular pathology, and α-synucleinopathy, which might contribute to either the pathogenesis or the clinical progression of AD, have also been discussed. Other emerging candidates including non-coding RNAs, metabolites, and extracellular vesicle-based markers have also enriched the biofluid biomarker landscape for AD. Moreover, the review discusses the current challenges of biofluid biomarkers in AD diagnosis and offers insights into the prospective future development.
Collapse
Affiliation(s)
- Chengyu An
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuiyue Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
22
|
Klinger HM, Healy BC, Hanseeuw BJ, Jones RN, Boyle R, Townsend DL, Properzi MJ, Coughlan GT, Seto M, Birkenbihl C, Farrell ME, Papp KV, Chhatwal JP, Yang HS, Schultz AP, Amariglio RE, Jacobs HIL, Price JC, Johnson KA, Rentz DM, Sperling RA, Buckley RF. Latent change-on-change between amyloid accumulation and cognitive decline. Alzheimers Dement 2024; 20:8728-8738. [PMID: 39470175 DOI: 10.1002/alz.14326] [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: 06/25/2024] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/30/2024]
Abstract
INTRODUCTION While the influence of cross-sectional β-amyloid (Aβ) on longitudinal changes in cognition is well established, longitudinal change-on-change between Aβ and cognition is less explored. METHODS A series of bivariate latent change score models (LCSM) examined the relationship between changes in 11C-Pittsburgh Compound-B (PiB) positron emission tomography (PET) and the Preclinical Alzheimer's Cognitive Composite-5 (PACC-5) while adjusting for covariates, including cross-sectional medial temporal lobe (MTL) tau-PET burden. We selected 352 clinically normal older participants with up to 9 years of PiB-PET and PACC-5 data from the Harvard Aging Brain Study (HABS). RESULTS Aβ accumulation was associated with subsequent cognitive decline beyond the effects of cross-sectional Aβ burden. Within this model including covariates such as age, sex, and apolipoprotein ε4 (APOEε4) status, we found no evidence supporting previously published associations between cross-sectional tau-PET and cognitive intercept/slope. DISCUSSION Short-term Aβ changes are significantly associated with cognitive decline in clinically normal older adults and may eclipse the effect of cross-sectional Aβ and MTL tau. HIGHLIGHTS Aβ accumulation is associated with subsequent cognitive decline. High Aβ burden is not the sole metric signaling impending cognitive decline. Contrary to prior work, MTL tau-PET and cognition were not associated in our models. Models of bivariate latent Aβ and cognitive change may eclipse the effects of MTL tau.
Collapse
Affiliation(s)
- Hannah M Klinger
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian C Healy
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bernard J Hanseeuw
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Cliniques Universitaires Saint-Luc, Institute of Neurosciences, UCLouvain, Belgium
| | - Rich N Jones
- Department of Psychiatry and Human Behavior, Brown University, Warren Alpert Medical School, Providence, Rhode Island, USA
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Diana L Townsend
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gillian T Coughlan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mabel Seto
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Colin Birkenbihl
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michelle E Farrell
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathryn V Papp
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Rebecca E Amariglio
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Heidi I L Jacobs
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Keith A Johnson
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dorene M Rentz
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Farhadieh ME, Mozafar M, Sanaaee S, Sodeifi P, Kousha K, Zare Y, Zare S, Maleki Rad N, Jamshidi-Goharrizi F, Allahverdloo M, Rahimi A, Sadeghi M, Shafie M, Mayeli M. Polygenic hazard score predicts synaptic and axonal degeneration and cognitive decline in Alzheimer's disease continuum. Arch Gerontol Geriatr 2024; 127:105576. [PMID: 39096557 DOI: 10.1016/j.archger.2024.105576] [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: 02/23/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Growth associated protein-43 (GAP-43) and neurofilaments light (NFL) are biomarkers of synaptic and axonal injury, and are associated with cognitive decline in Alzheimer's disease (AD) contiuum. We investigated whether Polygenic Hazard Score (PHS) is associated with specific biomarkers and cognitive measures, and if it can predict the relationship between GAP-43, NFL, and cognitive decline in AD. METHOD We enrolled 646 subjects: 93 with AD, 350 with mild cognitive impairment (MCI), and 203 cognitively normal controls. Variables included GAP-43, plasma NFL, and PHS. A PHS of 0.21 or higher was considered high risk while a PHS below this threshold was considered low risk. A subsample of 190 patients with MCI with four years of follow-up cognitive assessments were selected for longitudinal analysis . We assessed the association of the PHS with AD biomarkers and cognitive measures, as well as the predictive power of PHS on cognitive decline and the conversion of MCI to AD. RESULTS PHS showed high diagnostic accuracy in distinguishing AD, MCI, and controls. At each follow-up point, high risk MCI patients showed higher level of cognitive impairment compared to the low risk group. GAP-43 correlated with all follow-up cognitive tests in high risk MCI patients which was not detected in low risk MCI patients. Moreover, high risk MCI patients progressed to dementia more rapidly compared to low risk patients. CONCLUSION PHS can predict cognitive decline and impacts the relationship between neurodegenerative biomarkers and cognitive impairment in AD contiuum. Categorizing patients based on PHS can improve the prediction of cognitive outcomes and disease progression.
Collapse
Affiliation(s)
- Mohammad-Erfan Farhadieh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mehrdad Mozafar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; NeuroTRACT Association, Tehran University of Medical Sciences, Tehran, Iran
| | - Saameh Sanaaee
- Department of Cellular and Molecular Biology, School of Advanced Sciences and Technology, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Parastoo Sodeifi
- School of Medicine, Azad University of Medical Sciences, Tehran, Iran
| | - Kiana Kousha
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Yeganeh Zare
- Department of Psychology, Faculty of Literature, Humanities and Social Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shahab Zare
- Department of Educational Psychology, Faculty of Psychology and Educational Science, Allameh Tabatabai University, Tehran, Iran
| | - Nooshin Maleki Rad
- Department of Linguistic, Faculty of Literatures and Human Sciences, University of Ferdowsi, Mashhad, Iran
| | - Faezeh Jamshidi-Goharrizi
- Department of Sociology-Cultural Sociology, Islamic Azad University, Kish International Branch, Hormozgan, Iran; Department of Psychology, Faculty of Psychology, Payame Noor University, Tehran, Iran
| | - Mohammad Allahverdloo
- Department of Psychology, Faculty of Literature, Humanities and Social Sciences, Islamic Azad University, Zanjan, Iran
| | - Arman Rahimi
- Institue of Translational Medicine, Semmelwis, Budapest, Hungary
| | - Mohammad Sadeghi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; NeuroTRACT Association, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahan Shafie
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; NeuroTRACT Association, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Mayeli
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
24
|
Reyes RER, Wilson RE, Langhough RE, Studer RL, Jonaitis EM, Oomens JE, Planalp EM, Bendlin BB, Chin NA, Asthana S, Zetterberg H, Johnson SC. Targeted Proteomic Biomarker Profiling Using NULISA in a cohort enriched with risk for Alzheimer's Disease and Related Dementias. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.28.24318162. [PMID: 39649596 PMCID: PMC11623751 DOI: 10.1101/2024.11.28.24318162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
INTRODUCTION Targeted proteomic assays may be useful for diagnosing and staging Alzheimer's disease and related dementias (ADRD). We evaluated the performance of a 120-marker central nervous system (CNS) NUcleic acid-Linked Immuno-Sandwich Assay (NULISA) panel in samples spanning the AD spectrum. METHODS Cross-sectional plasma samples (n=252) were analyzed using Alamar's NULISAseq CNS panel. ROC analyses demonstrated NULISAseq-pTau217 accuracy in detecting amyloid (A) and tau (T) PET positivity. Differentially expressed proteins were identified using volcano plots. RESULTS NULISAseq-pTau217 accurately classified A/T PET status with ROC AUCs of 0.92/0.86. pTau217 was upregulated in A+, T+, and impaired groups with log2-fold changes of 1.21, 0.57 and 4.63, respectively, compared to A-. Interestingly, pTDP43-409 was also upregulated in the impaired group and correlated with declining hippocampal volume and cognitive trajectories. DISCUSSION This study shows the potential of a targeted proteomics panel for characterizing brain changes pertinent to ADRD. The promising pTDP43-409 findings require further replication.
Collapse
Affiliation(s)
- Ramiro Eduardo Rea Reyes
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Rachael E Wilson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, 610 Walnut Street, Madison, WI, 53726, USA
| | - Rachel L Studer
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, 610 Walnut Street, Madison, WI, 53726, USA
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, 610 Walnut Street, Madison, WI, 53726, USA
| | - Julie E Oomens
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Elizabeth M Planalp
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Nathaniel A Chin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Biskopsbogatan 27, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Biskopsbogatan 27, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, Gower Street, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Units 1501-1502, 1512-1518, 15/F Building 17W, 17 Science Park W Ave, Science Park, Hong Kong, 0000, China
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI, 53792, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, 610 Walnut Street, Madison, WI, 53726, USA
| |
Collapse
|
25
|
Miller AA, Sharp ES, Wang S, Zhao Y, Mecca AP, van Dyck CH, O'Dell RS. Self-reported hearing loss is associated with faster cognitive and functional decline but not diagnostic conversion in the ADNI cohort. Alzheimers Dement 2024; 20:7847-7858. [PMID: 39324520 PMCID: PMC11567835 DOI: 10.1002/alz.14252] [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: 04/18/2024] [Revised: 07/29/2024] [Accepted: 08/17/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Hearing loss is identified as one of the largest modifiable risk factors for cognitive impairment and dementia. Studies evaluating this relationship have yielded mixed results. METHODS We investigated the longitudinal relationship between self-reported hearing loss and cognitive/functional performance in 695 cognitively normal (CN) and 941 participants with mild cognitive impairment (MCI) enrolled in the Alzheimer's Disease Neuroimaging Initiative. RESULTS Within CN participants with hearing loss, there was a significantly greater rate of cognitive decline per modified preclinical Alzheimer's cognitive composite. Within both CN and MCI participants with hearing loss, there was a significantly greater rate of functional decline per the functional activities questionnaire (FAQ). In CN and MCI participants, hearing loss did not significantly contribute to the risk of progression to a more impaired diagnosis. DISCUSSION These results confirm previous studies demonstrating a significant longitudinal association between self-reported hearing loss and cognition/function but do not demonstrate an increased risk of conversion to a more impaired diagnosis. CLINICAL TRIAL REGISTRATION INFORMATION NCT00106899 (ADNI: Alzheimer's Disease Neuroimaging Initiative, clinicaltrials.gov), NCT01078636 (ADNI-GO: Alzheimer's Disease Neuroimaging Initiative Grand Opportunity, clinicaltrials.gov), NCT01231971 (ADNI2: Alzheimer's Disease Neuroimaging Initiative 2, clinicaltrials.gov), NCT02854033 (ADNI3: Alzheimer's Disease Neuroimaging Initiative 3, clinicaltrials.gov). HIGHLIGHTS Hearing loss is a potential modifiable risk factor for dementia. We assessed the effect of self-reported hearing loss on cognition and function in the ADNI cohort. Hearing loss contributes to significantly faster cognitive and functional decline. Hearing loss was not associated with conversion to a more impaired diagnosis.
Collapse
Grants
- P30AG021342 NIA NIH HHS
- GE Healthcare
- AbbVie, Alzheimer's Association
- P30AG066508 NIA NIH HHS
- Biogen; Bristol-Myers Squibb Company
- W81XWH-12-2-0012 Department of Defense
- EuroImmun
- Johnson & Johnson Pharmaceutical Research & Development LLC.
- Alzheimer's Drug Discovery Foundation
- UL1 TR001863 NCATS NIH HHS
- Servier
- Lumosity
- U01 AG024904 NIA NIH HHS
- Piramal Imaging
- Takeda Pharmaceutical Company
- P30 AG066508 NIA NIH HHS
- RF1 AG068191 NIA NIH HHS
- the Alzheimer's Disease Neuroimaging Initiative (ADNI)
- Araclon Biotech
- U01 AG024904 NIH HHS
- Novartis Pharmaceuticals Corporation
- Meso Scale Diagnostics, LLC.
- CereSpir, Inc.
- Northern California Institute for Research and Education
- BioClinica, Inc.
- RF1 AG081413 NIA NIH HHS
- P30 AG021342 NIA NIH HHS
- Transition Therapeutics
- Janssen Alzheimer Immunotherapy Research &Development, LLC.
- Cogstate; Eisai Inc.
- the National Institute of Biomedical Imaging and Bioengineering
- The Canadian Institutes of Health Research
- Pfizer Inc.
- Elan Pharmaceuticals, Inc.
- F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.
- Eli Lilly and Company
- IXICO Ltd.
- NeuroRx Research
- RF1AG081413 NIA NIH HHS
- Merck & Co., Inc.
- RF1AG068191 NIA NIH HHS
- Neurotrack Technologies
- Fujirebio
- Lundbeck
- National Institutes of Health
- Department of Defense
- National Institute on Aging
- Alzheimer's Drug Discovery Foundation
- BioClinica, Inc.
- Eli Lilly and Company
- Fujirebio
- GE Healthcare
- Lundbeck
- Merck & Co., Inc.
- Novartis Pharmaceuticals Corporation
- Pfizer Inc.
- Servier
- Takeda Pharmaceutical Company
- Northern California Institute for Research and Education
Collapse
Affiliation(s)
- Alyssa A. Miller
- Alzheimer's Disease Research UnitYale University School of MedicineNew HavenConnecticutUSA
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Emily S. Sharp
- Alzheimer's Disease Research UnitYale University School of MedicineNew HavenConnecticutUSA
- Department of NeurologyYale University School of MedicineNew HavenConnecticutUSA
| | - Selena Wang
- Department of BiostatisticsYale University School of Public HealthNew HavenConnecticutUSA
- Department of Biostatistics and Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yize Zhao
- Department of BiostatisticsYale University School of Public HealthNew HavenConnecticutUSA
| | - Adam P. Mecca
- Alzheimer's Disease Research UnitYale University School of MedicineNew HavenConnecticutUSA
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Christopher H. van Dyck
- Alzheimer's Disease Research UnitYale University School of MedicineNew HavenConnecticutUSA
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
- Department of NeurologyYale University School of MedicineNew HavenConnecticutUSA
- Department of NeuroscienceYale University School of MedicineNew HavenConnecticutUSA
| | - Ryan S. O'Dell
- Alzheimer's Disease Research UnitYale University School of MedicineNew HavenConnecticutUSA
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | | |
Collapse
|
26
|
Palmqvist S, Tideman P, Mattsson-Carlgren N, Schindler SE, Smith R, Ossenkoppele R, Calling S, West T, Monane M, Verghese PB, Braunstein JB, Blennow K, Janelidze S, Stomrud E, Salvadó G, Hansson O. Blood Biomarkers to Detect Alzheimer Disease in Primary Care and Secondary Care. JAMA 2024; 332:1245-1257. [PMID: 39068545 PMCID: PMC11284636 DOI: 10.1001/jama.2024.13855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024]
Abstract
Importance An accurate blood test for Alzheimer disease (AD) could streamline the diagnostic workup and treatment of AD. Objective To prospectively evaluate a clinically available AD blood test in primary care and secondary care using predefined biomarker cutoff values. Design, Setting, and Participants There were 1213 patients undergoing clinical evaluation due to cognitive symptoms who were examined between February 2020 and January 2024 in Sweden. The biomarker cutoff values had been established in an independent cohort and were applied to a primary care cohort (n = 307) and a secondary care cohort (n = 300); 1 plasma sample per patient was analyzed as part of a single batch for each cohort. The blood test was then evaluated prospectively in the primary care cohort (n = 208) and in the secondary care cohort (n = 398); 1 plasma sample per patient was sent for analysis within 2 weeks of collection. Exposure Blood tests based on plasma analyses by mass spectrometry to determine the ratio of plasma phosphorylated tau 217 (p-tau217) to non-p-tau217 (expressed as percentage of p-tau217) alone and when combined with the amyloid-β 42 and amyloid-β 40 (Aβ42:Aβ40) plasma ratio (the amyloid probability score 2 [APS2]). Main Outcomes and Measures The primary outcome was AD pathology (determined by abnormal cerebrospinal fluid Aβ42:Aβ40 ratio and p-tau217). The secondary outcome was clinical AD. The positive predictive value (PPV), negative predictive value (NPV), diagnostic accuracy, and area under the curve (AUC) values were calculated. Results The mean age was 74.2 years (SD, 8.3 years), 48% were women, 23% had subjective cognitive decline, 44% had mild cognitive impairment, and 33% had dementia. In both the primary care and secondary care assessments, 50% of patients had AD pathology. When the plasma samples were analyzed in a single batch in the primary care cohort, the AUC was 0.97 (95% CI, 0.95-0.99) when the APS2 was used, the PPV was 91% (95% CI, 87%-96%), and the NPV was 92% (95% CI, 87%-96%); in the secondary care cohort, the AUC was 0.96 (95% CI, 0.94-0.98) when the APS2 was used, the PPV was 88% (95% CI, 83%-93%), and the NPV was 87% (95% CI, 82%-93%). When the plasma samples were analyzed prospectively (biweekly) in the primary care cohort, the AUC was 0.96 (95% CI, 0.94-0.98) when the APS2 was used, the PPV was 88% (95% CI, 81%-94%), and the NPV was 90% (95% CI, 84%-96%); in the secondary care cohort, the AUC was 0.97 (95% CI, 0.95-0.98) when the APS2 was used, the PPV was 91% (95% CI, 87%-95%), and the NPV was 91% (95% CI, 87%-95%). The diagnostic accuracy was high in the 4 cohorts (range, 88%-92%). Primary care physicians had a diagnostic accuracy of 61% (95% CI, 53%-69%) for identifying clinical AD after clinical examination, cognitive testing, and a computed tomographic scan vs 91% (95% CI, 86%-96%) using the APS2. Dementia specialists had a diagnostic accuracy of 73% (95% CI, 68%-79%) vs 91% (95% CI, 88%-95%) using the APS2. In the overall population, the diagnostic accuracy using the APS2 (90% [95% CI, 88%-92%]) was not different from the diagnostic accuracy using the percentage of p-tau217 alone (90% [95% CI, 88%-91%]). Conclusions and Relevance The APS2 and percentage of p-tau217 alone had high diagnostic accuracy for identifying AD among individuals with cognitive symptoms in primary and secondary care using predefined cutoff values. Future studies should evaluate how the use of blood tests for these biomarkers influences clinical care.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Neurology Clinic, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Susanna Calling
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
- University Clinic Primary Care, Skåne, Sweden
| | - Tim West
- C2N Diagnostics LLC, St Louis, Missouri
| | | | | | | | - Kaj Blennow
- Paris Brain Institute, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - 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
| |
Collapse
|
27
|
Jagust WJ, Koeppe RA, Rabinovici GD, Villemagne VL, Harrison TM, Landau SM. 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
|
28
|
Beckett LA, Saito N, Donohue MC, Harvey DJ. Contributions of the ADNI Biostatistics Core. Alzheimers Dement 2024; 20:7331-7339. [PMID: 39140601 PMCID: PMC11485306 DOI: 10.1002/alz.14159] [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: 04/26/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 08/15/2024]
Abstract
The goal of the Biostatistics Core of the Alzheimer's Disease Neuroimaging Initiative (ADNI) has been to ensure that sound study designs and statistical methods are used to meet the overall goals of ADNI. We have supported the creation of a well-validated and well-curated longitudinal database of clinical and biomarker information on ADNI participants and helped to make this accessible and usable for researchers. We have developed a statistical methodology for characterizing the trajectories of clinical and biomarker change for ADNI participants across the spectrum from cognitively normal to dementia, including multivariate patterns and evidence for heterogeneity in cognitive aging. We have applied these methods and adapted them to improve clinical trial design. ADNI-4 will offer us a chance to help extend these efforts to a more diverse cohort with an even richer panel of biomarker data to support better knowledge of and treatment for Alzheimer's disease and related dementias. HIGHLIGHTS: The Alzheimer's Disease Neuroimaging Initiative (ADNI) Biostatistics Core provides study design and analytic support to ADNI investigators. Core members develop and apply novel statistical methodology to work with ADNI data and support clinical trial design. The Core contributes to the standardization, validation, and harmonization of biomarker data. The Core serves as a resource to the wider research community to address questions related to the data and study as a whole.
Collapse
Affiliation(s)
- Laurel A. Beckett
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Naomi Saito
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Michael C. Donohue
- Department of NeurologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Danielle J. Harvey
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | |
Collapse
|
29
|
Aisen PS, Donohue MC, Raman R, Rafii MS, Petersen RC. The Alzheimer's Disease Neuroimaging Initiative Clinical Core. Alzheimers Dement 2024; 20:7361-7368. [PMID: 39136045 PMCID: PMC11485391 DOI: 10.1002/alz.14167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 10/18/2024]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) Clinical Core is responsible for coordination of all clinical activities at the ADNI sites, including project management, regulatory oversight, and site management and monitoring, as well as the collection of all clinical data and management of all study data. The Clinical Core is also charged with determining the clinical classifications and criteria for enrollment in evolving AD trials and enabling the ongoing characterization of the cross-sectional features and longitudinal trajectories of the ADNI cohorts with application of these findings to optimal clinical trial designs. More than 2400 individuals have been enrolled in the cohorts since the inception of ADNI, facilitating refinement of our understanding of the AD trajectory and allowing academic and industry investigators to model therapeutic trials across the disease spectrum from the presymptomatic stage through dementia. HIGHLIGHTS: Since 2004, the Alzheimer's Disease Neuroimaging Initiative (ADNI) Clinical Core has overseen the enrollment of > 2400 participants with mild cognitive impairment, mild Alzheimer's disease (AD) dementia, and normal cognition. The longitudinal dataset has elucidated the full cognitive and clinical trajectory of AD from its presymptomatic stage through the onset of dementia. The ADNI data have supported the design of most major trials in the field.
Collapse
Affiliation(s)
- Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Michael C. Donohue
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Rema Raman
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | | | | |
Collapse
|
30
|
Vanderlip CR, Lee MD, Stark CEL. Cognitive modeling of the Mnemonic Similarity Task as a digital biomarker for Alzheimer's disease. Alzheimers Dement 2024; 20:6935-6947. [PMID: 39239893 PMCID: PMC11485396 DOI: 10.1002/alz.14163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND The Mnemonic Similarity Task (MST) is a popular memory task designed to assess hippocampal integrity. We assessed whether analyzing MST performance using a multinomial processing tree (MPT) cognitive model could detect individuals with elevated Alzheimer's disease (AD) biomarker status prior to cognitive decline. METHOD We analyzed MST data from >200 individuals (young, cognitively healthy older adults and individuals with mild cognitive impairment [MCI]), a subset of which also had existing cerebrospinal fluid (CSF) amyloid beta (Aβ) and phosphorylated tau (pTau) data using both traditional and model-derived approaches. We assessed how well each could predict age group, memory ability, MCI status, Aβ, and pTau status using receiver operating characteristic analyses. RESULTS Both approaches predicted age group membership equally, but MPT-derived metrics exceeded traditional metrics in all other comparisons. DISCUSSION A MPT model of the MST can detect individuals with AD prior to cognitive decline, making it a potentially useful tool for screening and monitoring older adults during the asymptomatic phase of AD. HIGHLIGHTS The MST, along with cognitive modeling, identifies individuals with memory deficits and cognitive impairment. Cognitive modeling of the MST identifies individuals with increased AD biomarkers prior to changes in cognitive function. The MST is a digital biomarker that identifies individuals at high risk of AD.
Collapse
Affiliation(s)
- Casey R. Vanderlip
- Department of Neurobiology and Behavior1424 Biological Sciences IIIUniversity of California, IrvineIrvineCaliforniaUSA
| | - Michael D. Lee
- Department of Cognitive Science3151 Social Sciences Plaza AUniversity of California, IrvineIrvineCaliforniaUSA
| | - Craig E. L. Stark
- Department of Neurobiology and Behavior1424 Biological Sciences IIIUniversity of California, IrvineIrvineCaliforniaUSA
| |
Collapse
|
31
|
Chou CJ, Chang CT, Chang YN, Lee CY, Chuang YF, Chiu YL, Liang WL, Fan YM, Liu YC. Screening for early Alzheimer's disease: enhancing diagnosis with linguistic features and biomarkers. Front Aging Neurosci 2024; 16:1451326. [PMID: 39376506 PMCID: PMC11456453 DOI: 10.3389/fnagi.2024.1451326] [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: 06/19/2024] [Accepted: 09/11/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction Research has shown that speech analysis demonstrates sensitivity in detecting early Alzheimer's disease (AD), but the relation between linguistic features and cognitive tests or biomarkers remains unclear. This study aimed to investigate how linguistic features help identify cognitive impairments in patients in the early stages of AD. Method This study analyzed connected speech from 80 participants and categorized the participants into early-AD and normal control (NC) groups. The participants underwent amyloid-β positron emission tomography scans, brain magnetic resonance imaging, and comprehensive neuropsychological testing. Participants' speech data from a picture description task were examined. A total of 15 linguistic features were analyzed to classify groups and predict cognitive performance. Results We found notable linguistic differences between the early-AD and NC groups in lexical diversity, syntactic complexity, and language disfluency. Using machine learning classifiers (SVM, KNN, and RF), we achieved up to 88% accuracy in distinguishing early-AD patients from normal controls, with mean length of utterance (MLU) and long pauses ratio (LPR) serving as core linguistic indicators. Moreover, the integration of linguistic indicators with biomarkers significantly improved predictive accuracy for AD. Regression analysis also highlighted crucial linguistic features, such as MLU, LPR, Type-to-Token ratio (TTR), and passive construction ratio (PCR), which were sensitive to changes in cognitive function. Conclusion Findings support the efficacy of linguistic analysis as a screening tool for the early detection of AD and the assessment of subtle cognitive decline. Integrating linguistic features with biomarkers significantly improved diagnostic accuracy.
Collapse
Affiliation(s)
- Chia-Ju Chou
- Department of Neurology, Cardinal Tien Hospital, Taipei, Taiwan
| | - Chih-Ting Chang
- Department of Speech-Language Pathology and Audiology, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Ya-Ning Chang
- Miin Wu School of Computing, National Cheng Kung University, Tainan, Taiwan
| | | | - Yi-Fang Chuang
- Institute of Public Health, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- International Health Program, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Health Innovation Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Ling Chiu
- Department of Medical Research, Far Eastern Memorial Hospital, Taipei, Taiwan
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Lin Liang
- Department of Neurology, Cardinal Tien Hospital, Taipei, Taiwan
| | - Yu-Ming Fan
- School of Medicine, Fu Jen Catholic University, Taipei, Taiwan
- Department of Nuclear Medicine, Cardinal Tien Hospital, Taipei, Taiwan
| | - Yi-Chien Liu
- Department of Neurology, Cardinal Tien Hospital, Taipei, Taiwan
- School of Medicine, Fu Jen Catholic University, Taipei, Taiwan
| |
Collapse
|
32
|
Collij LE, Mastenbroek SE, Mattsson-Carlgren N, Strandberg O, Smith R, Janelidze S, Palmqvist S, Ossenkoppele R, Hansson O. Lewy body pathology exacerbates brain hypometabolism and cognitive decline in Alzheimer's disease. Nat Commun 2024; 15:8061. [PMID: 39277604 PMCID: PMC11401923 DOI: 10.1038/s41467-024-52299-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024] Open
Abstract
Identifying concomitant Lewy body (LB) pathology through seed amplification assays (SAA) might enhance the diagnostic and prognostic work-up of Alzheimer's disease (AD) in clinical practice and trials. This study examined whether LB pathology exacerbates AD-related disease progression in 795 cognitively impaired individuals (Mild Cognitive Impairment and dementia) from the longitudinal multi-center observational ADNI cohort. Participants were on average 75 years of age (SD = 7.89), 40.8% were female, 184 (23.1%) had no biomarker evidence of AD/LB pathology, 39 (4.9%) had isolated LB pathology (AD-LB+), 395 (49.7%) had only AD pathology (AD+LB-), and 177 (22.3%) had both pathologies (AD+LB+). The AD+LB+ group showed worst baseline performance for most cognitive outcomes and compared to the AD+LB- group faster global cognitive decline and more cortical hypometabolism, particularly in posterior brain regions. Neuropathological examination (n = 61) showed high sensitivity (26/27, 96.3%) and specificity (27/28, 96.4%) of the SAA-test. We showed that co-existing LB-positivity exacerbates cognitive decline and cortical brain hypometabolism in AD. In vivo LB pathology detection could enhance prognostic evaluations in clinical practice and could have implications for clinical AD trial design.
Collapse
Affiliation(s)
- Lyduine E Collij
- Clinical memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands.
- Brain Imaging, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Sophie E Mastenbroek
- Clinical memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
- Brain Imaging, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Niklas Mattsson-Carlgren
- Clinical memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Olof Strandberg
- 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, Malmö, Sweden
| | - Shorena Janelidze
- Clinical memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Neurology, Alzheimercenter Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - 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.
| |
Collapse
|
33
|
Du L, Langhough RE, Wilson RE, Reyes RER, Hermann BP, Jonaitis EM, Betthauser TJ, Chin NA, Christian B, Chaby L, Jeromin A, Molfetta GD, Brum WS, Arslan B, Ashton N, Blennow K, Zetterberg H, Johnson SC. Longitudinal plasma phosphorylated-tau217 and other related biomarkers in a non-demented Alzheimer's risk-enhanced sample. Alzheimers Dement 2024; 20:6183-6204. [PMID: 38970274 PMCID: PMC11497664 DOI: 10.1002/alz.14100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 07/08/2024]
Abstract
INTRODUCTION Understanding longitudinal change in key plasma biomarkers will aid in detecting presymptomatic Alzheimer's disease (AD). METHODS Serial plasma samples from 424 Wisconsin Registry for Alzheimer's Prevention participants were analyzed for phosphorylated-tau217 (p-tau217; ALZpath) and other AD biomarkers, to study longitudinal trajectories in relation to disease, health factors, and cognitive decline. Of the participants, 18.6% with known amyloid status were amyloid positive (A+); 97.2% were cognitively unimpaired (CU). RESULTS In the CU, amyloid-negative (A-) subset, plasma p-tau217 levels increased modestly with age but were unaffected by body mass index and kidney function. In the whole sample, average p-tau217 change rates were higher in those who were A+ (e.g., simple slopes(se) for A+ and A- at age 60 were 0.232(0.028) and 0.038(0.013))). High baseline p-tau217 levels predicted faster preclinical cognitive decline. DISCUSSION p-tau217 stands out among markers for its strong association with disease and cognitive decline, indicating its potential for early AD detection and monitoring progression. HIGHLIGHTS Phosphorylated-tau217 (p-tau217) trajectories were significantly different in people who were known to be amyloid positive. Subtle age-related trajectories were seen for all the plasma markers in amyloid-negative cognitively unimpaired. Kidney function and body mass index were not associated with plasma p-tau217 trajectories. Higher plasma p-tau217 was associated with faster preclinical cognitive decline.
Collapse
Affiliation(s)
- Lianlian Du
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Rebecca E. Langhough
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Rachael E. Wilson
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Ramiro Eduardo Rea Reyes
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bruce P. Hermann
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of NeurologyUniversity of Wisconsin‐Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - Erin M. Jonaitis
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Tobey J. Betthauser
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Nathaniel A. Chin
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bradley Christian
- Waisman Laboratory for Brain Imaging and BehaviorUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | | | - Guglielmo Di Molfetta
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Wagner S. Brum
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Graduate Program in Biological Sciences: BiochemistryUniversidade Federal do Rio Grande do Sul (UFRGS)Porto AlegreRSBrazil
| | - Burak Arslan
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Nicholas Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- ICM Paris Brain Institute, ICMPitie‐Salpetriere HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiAnhuiChina
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongChina
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterDepartment of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| |
Collapse
|
34
|
Raket LL, Cummings J, Moscoso A, Villain N, Schöll M. Scenarios for the long-term efficacy of amyloid-targeting therapies in the context of the natural history of Alzheimer's disease. Alzheimers Dement 2024; 20:6374-6383. [PMID: 39073291 PMCID: PMC11497713 DOI: 10.1002/alz.14134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Recent clinical trials of amyloid beta (Aβ)-targeting therapies in Alzheimer's disease (AD) have demonstrated a clinical benefit over 18 months, but their long-term impact on disease trajectory is not yet understood. We propose a framework for evaluating realistic long-term scenarios. METHODS Results from recent phase 3 trials of Aβ-targeting antibodies were integrated with an estimate of the long-term patient-level natural history trajectory of the Clinical Dementia Rating-Sum of Boxes (CDR-SB) score to explore realistic long-term efficacy scenarios. RESULTS Three distinct long-term efficacy scenarios were examined, ranging from conservative to optimistic. These extrapolations of positive phase 3 trials suggested treatments delayed onset of severe dementia by 0.3 to 0.6 years (conservative), 1.1 to 1.9 years (intermediate), and 2.0 to 4.2 years (optimistic). DISCUSSION Our study provides a common language for long-term impact of disease-modifying treatments. Our work calls for studies with longer follow-up and results from early intervention trials to provide a comprehensive assessment of these therapies' true long-term impact. HIGHLIGHTS We present long-term scenarios of the efficacy of AD therapies. In this framework, scenarios are defined relative to the natural history of AD. Long-term projections with different levels of optimism can be compared. It provides a common language for expressing beliefs about long-term efficacy.
Collapse
Affiliation(s)
- Lars Lau Raket
- Eli Lilly and CompanyIndianapolisIndianaUSA
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
| | - Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeurosciencePam Quirk Brain Health and Biomarker LaboratoryDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Alexis Moscoso
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and NeurochemistryUniversity of GothenburgHuvudbyggnad Vasaparken, Universitetsplatsen 1GothenburgSweden
| | - Nicolas Villain
- Department of NeurologyInstitute of Memory and Alzheimer's DiseaseAP‐HP Sorbonne UniversitéPitié‐Salpêtrière HospitalParisFrance
- Sorbonne UniversitéINSERM U1127Institut du Cerveau ‐ ICMParisFrance
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and NeurochemistryUniversity of GothenburgHuvudbyggnad Vasaparken, Universitetsplatsen 1GothenburgSweden
- Department of Clinical PhysiologySahlgrenska University HospitalGothenburgSweden
- Dementia Research CentreQueen Square Institute of NeurologyUniversity College LondonLondonUK
| |
Collapse
|
35
|
Chen X, Dai Y, Li Y, Xin J, Zou J, Wang R, Zhang H, Liu Z. Identification of cross-talk pathways and PANoptosis-related genes in periodontitis and Alzheimer's disease by bioinformatics analysis and machine learning. Front Aging Neurosci 2024; 16:1430290. [PMID: 39258145 PMCID: PMC11384588 DOI: 10.3389/fnagi.2024.1430290] [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: 05/09/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
Background and objectives Periodontitis (PD), a chronic inflammatory disease, is a serious threat to oral health and is one of the risk factors for Alzheimer's disease (AD). A growing body of evidence suggests that the two diseases are closely related. However, current studies have not provided a comprehensive understanding of the common genes and common mechanisms between PD and AD. This study aimed to screen the crosstalk genes of PD and AD and the potential relationship between cross-talk and PANoptosis-related genes. The relationship between core genes and immune cells will be analyzed to provide new targets for clinical treatment. Materials and methods The PD and AD datasets were downloaded from the GEO database and differential expression analysis was performed to obtain DEGs. Overlapping DEGs had cross-talk genes linking PD and OP, and PANoptosis-related genes were obtained from a literature review. Pearson coefficients were used to compute cross-talk and PANoptosis-related gene correlations in the PD and AD datasets. Cross-talk genes were obtained from the intersection of PD and AD-related genes, protein-protein interaction(PPI) networks were constructed and cross-talk genes were identified using the STRING database. The intersection of cross-talk and PANoptosis-related genes was defined as cross-talk-PANoptosis genes. Core genes were screened using ROC analysis and XGBoost. PPI subnetwork, gene-biological process, and gene-pathway networks were constructed based on the core genes. In addition, immune infiltration on the PD and AD datasets was analyzed using the CIBERSORT algorithm. Results 366 cross-talk genes were overlapping between PD DEGs and AD DEGs. The intersection of cross-talk genes with 109 PANoptosis-related genes was defined as cross-talk-PANoptosis genes. ROC and XGBoost showed that MLKL, DCN, IL1B, and IL18 were more accurate than the other cross-talk-PANoptosis genes in predicting the disease, as well as better in overall characterization. GO and KEGG analyses showed that the four core genes were involved in immunity and inflammation in the organism. Immune infiltration analysis showed that B cells naive, Plasma cells, and T cells gamma delta were significantly differentially expressed in patients with PD and AD compared with the normal group. Finally, 10 drugs associated with core genes were retrieved from the DGIDB database. Conclusion This study reveals the joint mechanism between PD and AD associated with PANoptosis. Analyzing the four core genes and immune cells may provide new therapeutic directions for the pathogenesis of PD combined with AD.
Collapse
Affiliation(s)
- Xiantao Chen
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Yifei Dai
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Yushen Li
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Jiajun Xin
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Jiatong Zou
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Rui Wang
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Hao Zhang
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Zhihui Liu
- Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| |
Collapse
|
36
|
Vanderlip CR, Taylor L, Kim S, Harris AL, Tuteja N, Meza N, Escalante YY, McMillan L, Yassa MA, Adams JN. Amyloid-β deposition in basal frontotemporal cortex is associated with selective disruption of temporal mnemonic discrimination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609449. [PMID: 39253484 PMCID: PMC11383047 DOI: 10.1101/2024.08.23.609449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cerebral amyloid-beta (Aβ) accumulation, a hallmark pathology of Alzheimer's disease (AD), precedes clinical impairment by two to three decades. However, it is unclear whether Aβ contributes to subtle memory deficits observed during the preclinical stage. The heterogenous emergence of Aβ deposition may selectively impact certain memory domains, which rely on distinct underlying neural circuits. In this context, we tested whether specific domains of mnemonic discrimination, a neural computation essential for episodic memory, exhibit specific deficits related to early Aβ deposition. We tested 108 cognitively unimpaired human older adults (66% female) who underwent 18F-florbetapir positron emission tomography (Aβ-PET), and a control group of 35 young adults, on a suite of mnemonic discrimination tasks taxing object, spatial, and temporal domains. We hypothesized that Aβ pathology would be selectively associated with temporal discrimination performance due to Aβ's propensity to accumulate in the basal frontotemporal cortex, which supports temporal processing. Consistent with this hypothesis, we found a dissociation in which generalized age-related deficits were found for object and spatial mnemonic discrimination, while Aβ-PET levels were selectively associated with deficits in temporal mnemonic discrimination. Further, we found that higher Aβ-PET levels in medial orbitofrontal and inferior temporal cortex, regions supporting temporal processing, were associated with greater temporal mnemonic discrimination deficits, pointing to the selective vulnerability of circuits related to temporal processing early in AD progression. These results suggest that Aβ accumulation within basal frontotemporal regions may disrupt temporal mnemonic discrimination in preclinical AD, and may serve as a sensitive behavioral biomarker of emerging AD progression.
Collapse
Affiliation(s)
- Casey R. Vanderlip
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Lisa Taylor
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Soyun Kim
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Alyssa L. Harris
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Nandita Tuteja
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Novelle Meza
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Yuritza Y. Escalante
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Liv McMillan
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Michael A. Yassa
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| | - Jenna N. Adams
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697 USA
| |
Collapse
|
37
|
Wang J, Ackley S, Woodworth DC, Sajjadi SA, Decarli CS, Fletcher EF, Glymour MM, Jiang L, Kawas C, Corrada MM. Associations of Amyloid Burden, White Matter Hyperintensities, and Hippocampal Volume With Cognitive Trajectories in the 90+ Study. Neurology 2024; 103:e209665. [PMID: 39008782 PMCID: PMC11249511 DOI: 10.1212/wnl.0000000000209665] [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: 02/16/2024] [Accepted: 05/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Amyloid pathology, vascular disease pathology, and pathologies affecting the medial temporal lobe are associated with cognitive trajectories in older adults. However, only limited evidence exists on how these pathologies influence cognition in the oldest old. We evaluated whether amyloid burden, white matter hyperintensity (WMH) volume, and hippocampal volume (HV) are associated with cognitive level and decline in the oldest old. METHODS This was a longitudinal, observational community-based cohort study. We included participants with 18F-florbetapir PET and MRI data from the 90+ Study. Amyloid load was measured using the standardized uptake value ratio in the precuneus/posterior cingulate with eroded white matter mask as reference. WMH volume was log-transformed. All imaging measures were standardized using sample means and SDs. HV and log-WMH volume were normalized by total intracranial volume using the residual approach. Global cognitive performance was measured by the Mini-Mental State Examination (MMSE) and modified MMSE (3MS) tests, repeated every 6 months. We used linear mixed-effects models with random intercepts; random slopes; and interaction between time, time squared, and imaging variables to estimate the associations of imaging variables with cognitive level and cognitive decline. Models were adjusted for demographics, APOE genotype, and health behaviors. RESULTS The sample included 192 participants. The mean age was 92.9 years, 125 (65.1%) were female, 71 (37.0%) achieved a degree beyond college, and the median follow-up time was 3.0 years. A higher amyloid load was associated with a lower cognitive level (βMMSE = -0.82, 95% CI -1.17 to -0.46; β3MS = -2.77, 95% CI -3.69 to -1.84). A 1-SD decrease in HV was associated with a 0.70-point decrease in the MMSE score (95% CI -1.14 to -0.27) and a 2.27-point decrease in the 3MS score (95% CI -3.40 to -1.14). Clear nonlinear cognitive trajectories were detected. A higher amyloid burden and smaller HV were associated with faster cognitive decline. WMH volume was not significantly associated with cognitive level or decline. DISCUSSION Amyloid burden and hippocampal atrophy are associated with both cognitive level and cognitive decline in the oldest old. Our findings shed light on how different pathologies contributed to driving cognitive function in the oldest old.
Collapse
Affiliation(s)
- Jingxuan Wang
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Sarah Ackley
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Davis C Woodworth
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Seyed Ahmad Sajjadi
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Charles S Decarli
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Evan F Fletcher
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - M Maria Glymour
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Luohua Jiang
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Claudia Kawas
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Maria M Corrada
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| |
Collapse
|
38
|
Ni M, Zhu X, Wang K, Guo W, Shi Q, Li Y, Cui M, Xie Q. Novel β-amyloid PET Imaging Study of [ 18F]92 in Patients with Cognitive Decline. ACS OMEGA 2024; 9:34675-34683. [PMID: 39157119 PMCID: PMC11325415 DOI: 10.1021/acsomega.4c03412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024]
Abstract
[18F]-4-((E)-(((E)-4-(2-(2-(2-Fluoroethoxy)ethoxy)ethoxy)benzylidene)-hydrazono)methyl)-N-methylaniline ([18F]92) is a novel positron emission tomography (PET) tracer previously reported to exhibit high binding affinity to aggregated β-amyloid (Aβ). This study aims to report a fully automated radiosynthesis procedure for [18F]92, explore its radioactive distribution in the brains of healthy subjects, and investigate its potential application value in the early diagnosis of Alzheimer's disease (AD). The fully automated radiosynthesis of [18F]92 was performed on the AllinOne module. Thirty one participants were recruited for this study. Dynamic [18F]92 PET imaging was conducted over 0-90 min period to assess time-activity curves (TAC) and standardized uptake value ratio (SUVR) curves in cognitively normal (CN) subjects. All participants were visually classified as either positive (+) or negative (-). Semiquantitative analyses of [18F]92 were performed by calculating SUVRs in different regions of interest. Furthermore, the study analyzed the relationships between global SUVR and plasma AD biomarkers, including Aβ42, Aβ40, P-tau181, and T-tau. The automated radiosynthesis of [18F]92 was completed within 50 min, yielding a radiochemical purity of greater than 95% and a radiochemical yield of 36 ± 3% (nondecay-corrected). Among the participants, 15 were estimated as Aβ (-) and 16 as Aβ (+). TACs indicated that [18F]92 rapidly crossed the blood-brain barrier within 10 min, followed by a rapid decrease, which then slowed down in the last 50-90 min. SUVR curves revealed that SUVR values stabilized around 60-70 min after injection and reached an equilibrium between 70 and 90 min, primarily in the cerebral cortex. SUVRs of Aβ (+) participants were significantly higher than those of Aβ (-) individuals within the cerebral cortex. In addition, Aβ42 and the Aβ42/Aβ40 ratio exhibited negative correlations with global SUVR, while plasma P-tau181 and the P-tau181/T-tau ratio displayed positive correlations with global SUVR. [18F]92 exhibits excellent pharmacokinetic properties in the human brain and can be synthesized automatically on a large scale. [18F]92 is a promising and reliable radiotracer for estimating Aβ pathology accumulation, providing valuable assistance in AD diagnosis and guiding clinical trials of therapeutic drugs.
Collapse
Affiliation(s)
- Ming Ni
- Department
of Nuclear Medicine, the First Affiliated Hospital of USTC, Division
of Life Sciences and Medicine, University
of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xingxing Zhu
- Department
of Nuclear Medicine, the First Affiliated Hospital of USTC, Division
of Life Sciences and Medicine, University
of Science and Technology of China, Hefei, Anhui 230001, China
| | - Kaixuan Wang
- Department
of Nuclear Medicine, the First Affiliated Hospital of USTC, Division
of Life Sciences and Medicine, University
of Science and Technology of China, Hefei, Anhui 230001, China
- School
of Pharmacy, Bengbu Medical University, Bengbu 233000, China
| | - Wenliang Guo
- Department
of Neurology, the Second Hospital of Anhui
Medical University, Hefei, Anhui 230001, China
| | - Qin Shi
- Department
of Nuclear Medicine, the First Affiliated Hospital of USTC, Division
of Life Sciences and Medicine, University
of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yuying Li
- Key
Laboratory of Radiopharmaceuticals, Ministry of Education, College
of Chemistry, Beijing Normal University, Beijing 100875, China
- Center
for Advanced Materials Research, Beijing
Normal University at Zhuhai, Zhuhai 519087, China
| | - Mengchao Cui
- Key
Laboratory of Radiopharmaceuticals, Ministry of Education, College
of Chemistry, Beijing Normal University, Beijing 100875, China
- Center
for Advanced Materials Research, Beijing
Normal University at Zhuhai, Zhuhai 519087, China
| | - Qiang Xie
- Department
of Nuclear Medicine, the First Affiliated Hospital of USTC, Division
of Life Sciences and Medicine, University
of Science and Technology of China, Hefei, Anhui 230001, China
- School
of Pharmacy, Bengbu Medical University, Bengbu 233000, China
- Anhui
Provincial
Key Laboratory of Precision Pharmaceutical Preparations and Clinical
Pharmacy, Hefei, Anhui 230001, China
| |
Collapse
|
39
|
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
|
40
|
Wybitul M, Buchmann A, Langer N, Hock C, Treyer V, Gietl A. Trajectories of amyloid beta accumulation - Unveiling the relationship with APOE genotype and cognitive decline. Neurobiol Aging 2024; 139:44-53. [PMID: 38593527 DOI: 10.1016/j.neurobiolaging.2024.03.007] [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: 10/23/2023] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 04/11/2024]
Abstract
Amyloid beta (Aβ) follows a sigmoidal time function with varying accumulation rates. We studied how the position on this function, reflected by different Aβ accumulation phases, influences APOE ɛ4's association with Aβ and cognitive decline in 503 participants without dementia using Aβ-PET imaging over 5.3-years. First, Aβ load and accumulation were analyzed irrespective of phases using linear mixed regression. Generally, ɛ4 carriers displayed a higher Aβ load. Moreover, Aβ normal (Aβ-) ɛ4 carriers demonstrated higher accumulation. Next, we categorized accumulation phases as "decrease", "stable", or "increase" based on trajectory shapes. After excluding the Aβ-/decrease participants from the initial regression, the difference in accumulation attributable to genotype among Aβ- individuals was no longer significant. Further analysis revealed that in increase phases, Aβ accumulation was higher among noncarriers, indicating a genotype-related timeline shift. Finally, cognitive decline was analyzed across phases and was already evident in the Aβ-/increase phase. Our results encourage early interventions for ɛ4 carriers and imply that monitoring accumulating Aβ- individuals might help identify those at risk for cognitive decline.
Collapse
Affiliation(s)
- Maha Wybitul
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, Schlieren 8952, Switzerland; Department of Psychology, Faculty of Philosophy, University of Zurich, Zurich 8050, Switzerland
| | - Andreas Buchmann
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, Schlieren 8952, Switzerland
| | - Nicolas Langer
- Methods of Plasticity Research, Department of Psychology, University of Zurich, Zurich 8050, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, Schlieren 8952, Switzerland; Neurimmune, Schlieren 8952, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, Schlieren 8952, Switzerland; Department of Nuclear Medicine, University of Zurich, Zurich 8091, Switzerland.
| | - Anton Gietl
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, Schlieren 8952, Switzerland; University Hospital for Geriatric Psychiatry, Zurich 8008, Switzerland
| |
Collapse
|
41
|
Koychev I, Adler AI, Edison P, Tom B, Milton JE, Butchart J, Hampshire A, Marshall C, Coulthard E, Zetterberg H, Hellyer P, Cormack F, Underwood BR, Mummery CJ, Holman RR. Protocol for a double-blind placebo-controlled randomised controlled trial assessing the impact of oral semaglutide in amyloid positivity (ISAP) in community dwelling UK adults. BMJ Open 2024; 14:e081401. [PMID: 38908839 PMCID: PMC11328662 DOI: 10.1136/bmjopen-2023-081401] [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: 10/26/2023] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
INTRODUCTION Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), currently marketed for type 2 diabetes and obesity, may offer novel mechanisms to delay or prevent neurotoxicity associated with Alzheimer's disease (AD). The impact of semaglutide in amyloid positivity (ISAP) trial is investigating whether the GLP-1 RA semaglutide reduces accumulation in the brain of cortical tau protein and neuroinflammation in individuals with preclinical/prodromal AD. METHODS AND ANALYSIS ISAP is an investigator-led, randomised, double-blind, superiority trial of oral semaglutide compared with placebo. Up to 88 individuals aged ≥55 years with brain amyloid positivity as assessed by positron emission tomography (PET) or cerebrospinal fluid, and no or mild cognitive impairment, will be randomised. People with the low-affinity binding variant of the rs6971 allele of the Translocator Protein 18 kDa (TSPO) gene, which can interfere with interpreting TSPO PET scans (a measure of neuroinflammation), will be excluded.At baseline, participants undergo tau, TSPO PET and MRI scanning, and provide data on physical activity and cognition. Eligible individuals are randomised in a 1:1 ratio to once-daily oral semaglutide or placebo, starting at 3 mg and up-titrating to 14 mg over 8 weeks. They will attend safety visits and provide blood samples to measure AD biomarkers at weeks 4, 8, 26 and 39. All cognitive assessments are repeated at week 26. The last study visit will be at week 52, when all baseline measurements will be repeated. The primary end point is the 1-year change in tau PET signal. ETHICS AND DISSEMINATION The study was approved by the West Midlands-Edgbaston Research Ethics Committee (22/WM/0013). The results of the study will be disseminated through scientific presentations and peer-reviewed publications. TRIAL REGISTRATION NUMBER ISRCTN71283871.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Amanda I Adler
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Edison
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Brian Tom
- Medical Research Council Biostatistics Unit, University of Cambridge, UK
| | - Joanne E Milton
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Joe Butchart
- Royal Devon University Healthcare Foundation Trust, Exeter, UK
- University of Exeter Medical School, Exeter, UK
| | - Adam Hampshire
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Charles Marshall
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, People's Republic of China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA18 Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, UK
| | - Peter Hellyer
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | | | - Benjamin R Underwood
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation trust, Cambridge, UK
| | - Catherine J Mummery
- Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, UK
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
42
|
Quenon L, Collij LE, Garcia DV, Lopes Alves I, Gérard T, Malotaux V, Huyghe L, Gispert JD, Jessen F, Visser PJ, den Braber A, Ritchie CW, Boada M, Marquié M, Vandenberghe R, Luckett ES, Schöll M, Frisoni GB, Buckley C, Stephens A, Altomare D, Ford L, Birck C, Mett A, Gismondi R, Wolz R, Grootoonk S, Manber R, Shekari M, Lhommel R, Dricot L, Ivanoiu A, Farrar G, Barkhof F, Hanseeuw BJ. Amyloid-PET imaging predicts functional decline in clinically normal individuals. Alzheimers Res Ther 2024; 16:130. [PMID: 38886831 PMCID: PMC11181677 DOI: 10.1186/s13195-024-01494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND There is good evidence that elevated amyloid-β (Aβ) positron emission tomography (PET) signal is associated with cognitive decline in clinically normal (CN) individuals. However, it is less well established whether there is an association between the Aβ burden and decline in daily living activities in this population. Moreover, Aβ-PET Centiloids (CL) thresholds that can optimally predict functional decline have not yet been established. METHODS Cross-sectional and longitudinal analyses over a mean three-year timeframe were performed on the European amyloid-PET imaging AMYPAD-PNHS dataset that phenotypes 1260 individuals, including 1032 CN individuals and 228 participants with questionable functional impairment. Amyloid-PET was assessed continuously on the Centiloid (CL) scale and using Aβ groups (CL < 12 = Aβ-, 12 ≤ CL ≤ 50 = Aβ-intermediate/Aβ± , CL > 50 = Aβ+). Functional abilities were longitudinally assessed using the Clinical Dementia Rating (Global-CDR, CDR-SOB) and the Amsterdam Instrumental Activities of Daily Living Questionnaire (A-IADL-Q). The Global-CDR was available for the 1260 participants at baseline, while baseline CDR-SOB and A-IADL-Q scores and longitudinal functional data were available for different subsamples that had similar characteristics to those of the entire sample. RESULTS Participants included 765 Aβ- (61%, Mdnage = 66.0, IQRage = 61.0-71.0; 59% women), 301 Aβ± (24%; Mdnage = 69.0, IQRage = 64.0-75.0; 53% women) and 194 Aβ+ individuals (15%, Mdnage = 73.0, IQRage = 68.0-78.0; 53% women). Cross-sectionally, CL values were associated with CDR outcomes. Longitudinally, baseline CL values predicted prospective changes in the CDR-SOB (bCL*Time = 0.001/CL/year, 95% CI [0.0005,0.0024], p = .003) and A-IADL-Q (bCL*Time = -0.010/CL/year, 95% CI [-0.016,-0.004], p = .002) scores in initially CN participants. Increased clinical progression (Global-CDR > 0) was mainly observed in Aβ+ CN individuals (HRAβ+ vs Aβ- = 2.55, 95% CI [1.16,5.60], p = .020). Optimal thresholds for predicting decline were found at 41 CL using the CDR-SOB (bAβ+ vs Aβ- = 0.137/year, 95% CI [0.069,0.206], p < .001) and 28 CL using the A-IADL-Q (bAβ+ vs Aβ- = -0.693/year, 95% CI [-1.179,-0.208], p = .005). CONCLUSIONS Amyloid-PET quantification supports the identification of CN individuals at risk of functional decline. TRIAL REGISTRATION The AMYPAD PNHS is registered at www.clinicaltrialsregister.eu with the EudraCT Number: 2018-002277-22.
Collapse
Affiliation(s)
- Lisa Quenon
- Institute of Neuroscience, UCLouvain, Brussels, Belgium.
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - David Vállez Garcia
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Isadora Lopes Alves
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Brain Research Center, Amsterdam, The Netherlands
| | - Thomas Gérard
- Institute of Neuroscience, UCLouvain, Brussels, Belgium
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Vincent Malotaux
- Institute of Neuroscience, UCLouvain, Brussels, Belgium
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Lara Huyghe
- Institute of Neuroscience, UCLouvain, Brussels, Belgium
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands
| | - Anouk den Braber
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Craig W Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center for Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center for Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Louvain, Belgium
- Neurology Service, University Hospital Leuven, Louvain, Belgium
| | - Emma S Luckett
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Louvain, Belgium
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Göteborg, Sweden
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, University Hospital of Geneva, Geneva, Switzerland
| | | | | | - Daniele Altomare
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Lisa Ford
- Johnson & Johnson Innovative Medicine, Titusville, NJ, USA
| | | | - Anja Mett
- GE HealthCare, Glattbrugg, Switzerland
| | | | | | | | | | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Renaud Lhommel
- Institute of Neuroscience, UCLouvain, Brussels, Belgium
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | | | - Adrian Ivanoiu
- Institute of Neuroscience, UCLouvain, Brussels, Belgium
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | | | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Bernard J Hanseeuw
- Institute of Neuroscience, UCLouvain, Brussels, Belgium
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Gordon Center for Medical Imaging, Department of Radiology, Mass General Brigham, Boston, MA, USA
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
43
|
Cody KA, Langhough RE, Zammit MD, Clark L, Chin N, Christian BT, Betthauser TJ, Johnson SC. Characterizing brain tau and cognitive decline along the amyloid timeline in Alzheimer's disease. Brain 2024; 147:2144-2157. [PMID: 38667631 PMCID: PMC11146417 DOI: 10.1093/brain/awae116] [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: 10/30/2023] [Revised: 02/23/2024] [Accepted: 03/24/2024] [Indexed: 06/04/2024] Open
Abstract
Recent longitudinal PET imaging studies have established methods to estimate the age at which amyloid becomes abnormal at the level of the individual. Here we recontextualized amyloid levels into the temporal domain to better understand the downstream Alzheimer's disease processes of tau neurofibrillary tangle (NFT) accumulation and cognitive decline. This cohort study included a total of 601 individuals from the Wisconsin Registry for Alzheimer's Prevention and Wisconsin Alzheimer's Disease Research Center that underwent amyloid and tau PET, longitudinal neuropsychological assessments and met clinical criteria for three clinical diagnosis groups: cognitively unimpaired (n = 537); mild cognitive impairment (n = 48); or dementia (n = 16). Cortical 11C-Pittsburgh compound B (PiB) distribution volume ratio (DVR) and sampled iterative local approximation were used to estimate amyloid positive (A+; global PiB DVR > 1.16 equivalent to 17.1 centiloids) onset age and years of A+ duration at tau PET (i.e. amyloid chronicity). Tau PET burden was quantified using 18F-MK-6240 standardized uptake value ratios (70-90 min, inferior cerebellar grey matter reference region). Whole-brain and region-specific approaches were used to examine tau PET binding along the amyloid timeline and across the Alzheimer's disease clinical continuum. Voxel-wise 18F-MK-6240 analyses revealed that with each decade of A+, the spatial extent of measurable tau spread (i.e. progressed) from regions associated with early to late NFT tau stages. Regional analyses indicated that tau burden in the entorhinal cortex was detectable, on average, within 10 years of A+ onset. Additionally, the entorhinal cortex was the region most sensitive to early amyloid pathology and clinical impairment in this predominantly preclinical sample. Among initially cognitively unimpaired (n = 472) individuals with longitudinal cognitive follow-up, mixed effects models showed significant linear and non-linear interactions of A+ duration and entorhinal tau on cognitive decline, suggesting a synergistic effect whereby greater A+ duration, together with a higher entorhinal tau burden, increases the likelihood of cognitive decline beyond their separable effects. Overall, the amyloid time framework enabled a spatiotemporal characterization of tau deposition patterns across the Alzheimer's disease continuum. This approach, which examined cross-sectional tau PET data along the amyloid timeline to make longitudinal disease course inferences, demonstrated that A+ duration explains a considerable amount of variability in the magnitude and topography of tau spread, which largely recapitulated NFT staging observed in human neuropathological studies. By anchoring disease progression to the onset of amyloid, this study provides a temporal disease context, which may help inform disease prognosis and timing windows for anti-amyloid therapies.
Collapse
Affiliation(s)
- Karly A Cody
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Matthew D Zammit
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Lindsay Clark
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Nathaniel Chin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Bradley T Christian
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| |
Collapse
|
44
|
Mendes AJ, Ribaldi F, Lathuiliere A, Ashton NJ, Zetterberg H, Abramowicz M, Scheffler M, Assal F, Garibotto V, Blennow K, Frisoni GB. Comparison of plasma and neuroimaging biomarkers to predict cognitive decline in non-demented memory clinic patients. Alzheimers Res Ther 2024; 16:110. [PMID: 38755703 PMCID: PMC11097559 DOI: 10.1186/s13195-024-01478-9] [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: 02/05/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Plasma biomarkers of Alzheimer's disease (AD) pathology, neurodegeneration, and neuroinflammation are ideally suited for secondary prevention programs in self-sufficient persons at-risk of dementia. Plasma biomarkers have been shown to be highly correlated with traditional imaging biomarkers. However, their comparative predictive value versus traditional AD biomarkers is still unclear in cognitively unimpaired (CU) subjects and with mild cognitive impairment (MCI). METHODS Plasma (Aβ42/40, p-tau181, p-tau231, NfL, and GFAP) and neuroimaging (hippocampal volume, centiloid of amyloid-PET, and tau-SUVR of tau-PET) biomarkers were assessed at baseline in 218 non-demented subjects (CU = 140; MCI = 78) from the Geneva Memory Center. Global cognition (MMSE) was evaluated at baseline and at follow-ups up to 5.7 years. We used linear mixed-effects models and Cox proportional-hazards regression to assess the association between biomarkers and cognitive decline. Lastly, sample size calculations using the linear mixed-effects models were performed on subjects positive for amyloid-PET combined with tau-PET and plasma biomarker positivity. RESULTS Cognitive decline was significantly predicted in MCI by baseline plasma NfL (β=-0.55), GFAP (β=-0.36), hippocampal volume (β = 0.44), centiloid (β=-0.38), and tau-SUVR (β=-0.66) (all p < 0.05). Subgroup analysis with amyloid-positive MCI participants also showed that only NfL and GFAP were the only significant predictors of cognitive decline among plasma biomarkers. Overall, NfL and tau-SUVR showed the highest prognostic values (hazard ratios of 7.3 and 5.9). Lastly, we demonstrated that adding NfL to the inclusion criteria could reduce the sample sizes of future AD clinical trials by up to one-fourth in subjects with amyloid-PET positivity or by half in subjects with amyloid-PET and tau-PET positivity. CONCLUSIONS Plasma NfL and GFAP predict cognitive decline in a similar manner to traditional imaging techniques in amyloid-positive MCI patients. Hence, even though they are non-specific biomarkers of AD, both can be implemented in memory clinic workups as important prognostic biomarkers. Likewise, future clinical trials might employ plasma biomarkers as additional inclusion criteria to stratify patients at higher risk of cognitive decline to reduce sample sizes and enhance effectiveness.
Collapse
Affiliation(s)
- Augusto J Mendes
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.
- Geneva Memory Center, Department of Rehabilitation and Geriatrics,, Geneva University Hospitals, Geneva, Switzerland.
| | - Federica 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
| | - Aurelien Lathuiliere
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics,, Geneva University Hospitals, Geneva, Switzerland
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at 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, WI, USA
| | - Marc Abramowicz
- Genetic Medicine, Diagnostics Dept, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - Frédéric Assal
- Division of Neurology, Department of Clinical Neurosciences, Faculty of Medicine, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Faculty of Medicine, Geneva University Neurocenter, University of Geneva, Geneva, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland
- CIBM Center for Biomedical Imaging, Geneva, Switzerland
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Giovanni 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
|
45
|
Liu Z, Shi D, Cai Y, Li A, Lan G, Sun P, Liu L, Zhu Y, Yang J, Zhou Y, Guo L, Zhang L, Deng S, Chen S, Yu X, Chen X, Zhao R, Wang Q, Ran P, Xu L, Zhou L, Sun K, Wang X, Peng Q, Han Y, Guo T. Pathophysiology characterization of Alzheimer's disease in South China's aging population: for the Greater-Bay-Area Healthy Aging Brain Study (GHABS). Alzheimers Res Ther 2024; 16:84. [PMID: 38627753 PMCID: PMC11020808 DOI: 10.1186/s13195-024-01458-z] [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: 10/28/2023] [Accepted: 04/12/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION The Guangdong-Hong Kong-Macao Greater-Bay-Area of South China has an 86 million population and faces a significant challenge of Alzheimer's disease (AD). However, the characteristics and prevalence of AD in this area are still unclear due to the rarely available community-based neuroimaging AD cohort. METHODS Following the standard protocols of the Alzheimer's Disease Neuroimaging Initiative, the Greater-Bay-Area Healthy Aging Brain Study (GHABS) was initiated in 2021. GHABS participants completed clinical assessments, plasma biomarkers, genotyping, magnetic resonance imaging (MRI), β-amyloid (Aβ) positron emission tomography (PET) imaging, and tau PET imaging. The GHABS cohort focuses on pathophysiology characterization and early AD detection in the Guangdong-Hong Kong-Macao Greater Bay Area. In this study, we analyzed plasma Aβ42/Aβ40 (A), p-Tau181 (T), neurofilament light, and GFAP by Simoa in 470 Chinese older adults, and 301, 195, and 70 had MRI, Aβ PET, and tau PET, respectively. Plasma biomarkers, Aβ PET, tau PET, hippocampal volume, and temporal-metaROI cortical thickness were compared between normal control (NC), subjective cognitive decline (SCD), mild cognitive impairment (MCI), and dementia groups, controlling for age, sex, and APOE-ε4. The prevalence of plasma A/T profiles and Aβ PET positivity were also determined in different diagnostic groups. RESULTS The aims, study design, data collection, and potential applications of GHABS are summarized. SCD individuals had significantly higher plasma p-Tau181 and plasma GFAP than the NC individuals. MCI and dementia patients showed more abnormal changes in all the plasma and neuroimaging biomarkers than NC and SCD individuals. The frequencies of plasma A+/T+ (NC; 5.9%, SCD: 8.2%, MCI: 25.3%, dementia: 64.9%) and Aβ PET positivity (NC: 25.6%, SCD: 22.5%, MCI: 47.7%, dementia: 89.3%) were reported. DISCUSSION The GHABS cohort may provide helpful guidance toward designing standard AD community cohorts in South China. This study, for the first time, reported the pathophysiology characterization of plasma biomarkers, Aβ PET, tau PET, hippocampal atrophy, and AD-signature cortical thinning, as well as the prevalence of Aβ PET positivity in the Guangdong-Hong Kong-Macao Greater Bay Area of China. These findings provide novel insights into understanding the characteristics of abnormal AD pathological changes in South China's older population.
Collapse
Affiliation(s)
- Zhen Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Dai Shi
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Yue Cai
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Anqi Li
- 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
| | - Pan Sun
- 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
| | - Yalin Zhu
- 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
| | - Yajing Zhou
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Lizhi Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Laihong Zhang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Shuqing Deng
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Shuda Chen
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Xianfeng Yu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xuhui Chen
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Ruiyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Qingyong Wang
- Department of Neurology, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518107, China
| | - Pengcheng Ran
- Department of Nuclear Medicine, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Linsen Xu
- Department of Medical Imaging, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518106, China
| | - Liemin Zhou
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xinlu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Qiyu Peng
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, 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, 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
| | - 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.
| |
Collapse
|
46
|
Fujishima M, Kawasaki Y, Mitsuhashi T, Matsuda H. Impact of amyloid and tau positivity on longitudinal brain atrophy in cognitively normal individuals. Alzheimers Res Ther 2024; 16:77. [PMID: 38600602 PMCID: PMC11005141 DOI: 10.1186/s13195-024-01450-7] [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: 10/19/2023] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Individuals on the preclinical Alzheimer's continuum, particularly those with both amyloid and tau positivity (A + T +), display a rapid cognitive decline and elevated disease progression risk. However, limited studies exist on brain atrophy trajectories within this continuum over extended periods. METHODS This study involved 367 ADNI participants grouped based on combinations of amyloid and tau statuses determined through cerebrospinal fluid tests. Using longitudinal MRI scans, brain atrophy was determined according to the whole brain, lateral ventricle, and hippocampal volumes and cortical thickness in AD-signature regions. Cognitive performance was evaluated with the Preclinical Alzheimer's Cognitive Composite (PACC). A generalized linear mixed-effects model was used to examine group × time interactions for these measures. In addition, progression risks to mild cognitive impairment (MCI) or dementia were compared among the groups using Cox proportional hazards models. RESULTS A total of 367 participants (48 A + T + , 86 A + T - , 63 A - T + , and 170 A - T - ; mean age 73.8 years, mean follow-up 5.1 years, and 47.4% men) were included. For the lateral ventricle and PACC score, the A + T - and A + T + groups demonstrated statistically significantly greater volume expansion and cognitive decline over time than the A - T - group (lateral ventricle: β = 0.757 cm3/year [95% confidence interval 0.463 to 1.050], P < .001 for A + T - , and β = 0.889 cm3/year [0.523 to 1.255], P < .001 for A + T + ; PACC: β = - 0.19 /year [- 0.36 to - 0.02], P = .029 for A + T - , and β = - 0.59 /year [- 0.80 to - 0.37], P < .001 for A + T +). Notably, the A + T + group exhibited additional brain atrophy including the whole brain (β = - 2.782 cm3/year [- 4.060 to - 1.504], P < .001), hippocampus (β = - 0.057 cm3/year [- 0.085 to - 0.029], P < .001), and AD-signature regions (β = - 0.02 mm/year [- 0.03 to - 0.01], P < .001). Cox proportional hazards models suggested an increased risk of progressing to MCI or dementia in the A + T + group versus the A - T - group (adjusted hazard ratio = 3.35 [1.76 to 6.39]). CONCLUSIONS In cognitively normal individuals, A + T + compounds brain atrophy and cognitive deterioration, amplifying the likelihood of disease progression. Therapeutic interventions targeting A + T + individuals could be pivotal in curbing brain atrophy, cognitive decline, and disease progression.
Collapse
Affiliation(s)
- Motonobu Fujishima
- Department of Radiology, Kumagaya General Hospital, 4-5-1 Nakanishi, Kumagaya, 360-8567, Japan.
| | - Yohei Kawasaki
- Department of Biostatistics, Graduate School of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, 350-0495, Japan
- Biostatistics Section, Clinical Research Center, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8670, Japan
| | - Toshiharu Mitsuhashi
- Center for Innovative Clinical Medicine, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Hiroshi Matsuda
- Department of Biofunctional Imaging, Fukushima Medical University, 1 Hikariga-Oka, Fukushima, 960-1295, Japan
- Drug Discovery and Cyclotron Research Center, Southern Tohoku Research Institute for Neuroscience, 7-61-2 Yatsuyamada, Koriyama, 963-8052, Japan
| |
Collapse
|
47
|
Cui L, Zhang Z, Guo Y, Li Y, Xie F, Guo Q. Category Switching Test: A Brief Amyloid-β-Sensitive Assessment Tool for Mild Cognitive Impairment. Assessment 2024; 31:543-556. [PMID: 37081801 DOI: 10.1177/10731911231167537] [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: 04/22/2023]
Abstract
The Category Switching Test (CaST) is a verbal fluency test with active semantic category switching. This study aimed to explore the association between CaST performance and brain amyloid-β (Aβ) burden in patients with mild cognitive impairment (MCI) and the neurofunctional mechanisms. A total of 112 participants with MCI underwent Florbetapir positron emission tomography, resting-state functional magnetic resonance imaging, and a neuropsychological test battery. The high Aβ burden group had worse CaST performance than the low-burden group. CaST score and left middle temporal gyrus fractional amplitude of low-frequency fluctuations (fALFF) related inversely to the global Florbetapir standardized uptake value rate. Functional connectivity between the left middle temporal gyrus and frontal lobe decreased widely and correlated with CaST score in the high Aβ burden group. Thus, CaST score and left middle temporal gyrus fALFF were valuable in discriminating high Aβ burden. CaST might be useful in screening for MCI with high Aβ burden.
Collapse
Affiliation(s)
- Liang Cui
- Department of Gerontology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Zhang
- Department of Gerontology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihan Guo
- School of Medicine, The University of Queensland, Brisbane, Australia
| | - Yuehua Li
- Department of Gerontology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Barthélemy NR, Salvadó G, Schindler SE, He Y, Janelidze S, Collij LE, Saef B, Henson RL, Chen CD, Gordon BA, Li Y, La Joie R, Benzinger TLS, Morris JC, Mattsson-Carlgren N, Palmqvist S, Ossenkoppele R, Rabinovici GD, Stomrud E, Bateman RJ, Hansson O. Highly accurate blood test for Alzheimer's disease is similar or superior to clinical cerebrospinal fluid tests. Nat Med 2024; 30:1085-1095. [PMID: 38382645 PMCID: PMC11031399 DOI: 10.1038/s41591-024-02869-z] [Citation(s) in RCA: 133] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/12/2024] [Indexed: 02/23/2024]
Abstract
With the emergence of Alzheimer's disease (AD) disease-modifying therapies, identifying patients who could benefit from these treatments becomes critical. In this study, we evaluated whether a precise blood test could perform as well as established cerebrospinal fluid (CSF) tests in detecting amyloid-β (Aβ) plaques and tau tangles. Plasma %p-tau217 (ratio of phosporylated-tau217 to non-phosphorylated tau) was analyzed by mass spectrometry in the Swedish BioFINDER-2 cohort (n = 1,422) and the US Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC) cohort (n = 337). Matched CSF samples were analyzed with clinically used and FDA-approved automated immunoassays for Aβ42/40 and p-tau181/Aβ42. The primary and secondary outcomes were detection of brain Aβ or tau pathology, respectively, using positron emission tomography (PET) imaging as the reference standard. Main analyses were focused on individuals with cognitive impairment (mild cognitive impairment and mild dementia), which is the target population for available disease-modifying treatments. Plasma %p-tau217 was clinically equivalent to FDA-approved CSF tests in classifying Aβ PET status, with an area under the curve (AUC) for both between 0.95 and 0.97. Plasma %p-tau217 was generally superior to CSF tests in classification of tau-PET with AUCs of 0.95-0.98. In cognitively impaired subcohorts (BioFINDER-2: n = 720; Knight ADRC: n = 50), plasma %p-tau217 had an accuracy, a positive predictive value and a negative predictive value of 89-90% for Aβ PET and 87-88% for tau PET status, which was clinically equivalent to CSF tests, further improving to 95% using a two-cutoffs approach. Blood plasma %p-tau217 demonstrated performance that was clinically equivalent or superior to clinically used FDA-approved CSF tests in the detection of AD pathology. Use of high-performance blood tests in clinical practice can improve access to accurate AD diagnosis and AD-specific treatments.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lyduine E Collij
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Benjamin Saef
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel L Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Gil D Rabinovici
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA.
| | - 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.
| |
Collapse
|
49
|
Fonseca CS, Baker SL, Dobyns L, Janabi M, Jagust WJ, Harrison TM. Tau accumulation and atrophy predict amyloid independent cognitive decline in aging. Alzheimers Dement 2024; 20:2526-2537. [PMID: 38334195 PMCID: PMC11032527 DOI: 10.1002/alz.13654] [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: 08/24/2023] [Revised: 11/15/2023] [Accepted: 11/30/2023] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Amyloid beta (Aβ) and tau pathology are cross-sectionally associated with atrophy and cognitive decline in aging and Alzheimer's disease (AD). METHODS We investigated relationships between concurrent longitudinal measures of Aβ (Pittsburgh compound B [PiB] positron emission tomography [PET]), tau (flortaucipir [FTP] PET), atrophy (structural magnetic resonance imaging), episodic memory (EM), and non-memory (NM) in 78 cognitively healthy older adults (OA). RESULTS Entorhinal FTP change was correlated with EM decline regardless of Aβ, but meta-temporal FTP and global PiB change were only associated with EM and NM decline in Aβ+ OA. Voxel-wise analyses revealed significant associations between temporal lobe FTP change and EM decline in all groups. PiB and FTP change were not associated with structural change, suggesting a functional or microstructural mechanism linking these measures to cognitive decline. DISCUSSION Our results show that longitudinal Aβ is linked to cognitive decline only in the presence of elevated Aβ, but longitudinal temporal lobe tau is associated with memory decline regardless of Aβ status. HIGHLIGHTS Entorhinal tau change was associated with memory decline in older adults (OA), regardless of amyloid beta (Aβ). Greater meta-region of interest (ROI) tau change correlated with memory decline in Aβ+ OA. Voxel-wise temporal tau change correlated with memory decline, regardless of Aβ. Meta-ROI tau and global amyloid change correlated with non-memory change in Aβ+ OA. Tau and amyloid accumulation were not associated with structural change in OA.
Collapse
Affiliation(s)
- Corrina S. Fonseca
- Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | | - Lindsey Dobyns
- Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Mustafa Janabi
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - William J. Jagust
- Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Theresa M. Harrison
- Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| |
Collapse
|
50
|
Ackley S, Calmasini C, Bouteloup V, Hill-Jarrett TG, Swinnerton KN, Chêne G, Dufouil C, Glymour MM. Contribution of Global Amyloid-PET Imaging for Predicting Future Cognition in the MEMENTO Cohort. Neurology 2024; 102:e208054. [PMID: 38412412 PMCID: PMC11770678 DOI: 10.1212/wnl.0000000000208054] [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: 03/01/2023] [Accepted: 10/16/2023] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Global amyloid-PET is associated with cognition and cognitive decline, but most research on this association does not account for past cognitive information. We assessed the prognostic benefit of amyloid-PET measures for future cognition when prior cognitive assessments are available, evaluating the added value of amyloid measures beyond information on multiple past cognitive assessments. METHODS The French MEMENTO cohort (a cohort of outpatients from French research memory centers to improve knowledge on Alzheimer disease and related disorders) includes older outpatients with incipient cognitive changes, but no dementia diagnosis at inclusion. Global amyloid burden was assessed using positron emission tomography (amyloid-PET) for a subset of participants; semiannual cognitive testing was subsequently performed. We predicted mini-mental state examination (MMSE) scores using demographic characteristics (age, sex, marital status, and education) alone or in combination with information on prior cognitive measures. The added value of amyloid burden as a predictor in these models was evaluated with percent reduction of the mean squared error (MSE). All models were conducted separately for evaluating the added value of dichotomous amyloid positivity status compared with a continuous amyloid-standardized uptake-value ratio. RESULTS Our analytic sample comprised 510 individuals who underwent amyloid-PET scans with at least 4 MMSE assessments. The mean age at the PET scan was 71.6 (standard deviation 7.4) years; 60.7% were female. The median follow-up was 4.6 years (interquartile range: 0.9 years). Adding amyloid burden when adjusting for only demographic characteristics reduced the MSE of predictions by 5.08% (95% CI 0.97%-10.86%) and 12.64% (95% CI 3.35%-25.28%) for binary and continuous amyloid, respectively. If the model included 1 past MMSE measure, the MSE improvement was 3.51% (95% CI 1.01%-7.28%) when adding binary amyloid and 8.83% (95% CI 2.63%-16.37%) when adding continuous amyloid. Improvements in model fit were smaller with the addition of amyloid burden when more than 1 past cognitive assessment was included. For all models incorporating past cognitive assessments, differences in predictions amounted to a fraction of 1 MMSE point on average. DISCUSSION In a clinical setting, global amyloid burden did not appreciably improve cognitive predictions when past cognitive assessments were available. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT02164643.
Collapse
Affiliation(s)
- Sarah Ackley
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Camilla Calmasini
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Vincent Bouteloup
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Tanisha G Hill-Jarrett
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Kaitlin N Swinnerton
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Geneviève Chêne
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - Carole Dufouil
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| | - M M Glymour
- From the Department of Epidemiology (S.A., M.M.G.), Boston University, MA; Department of Epidemiology and Biostatistics (C.C., K.N.S.), University of California, San Francisco; University Bordeaux (V.B., G.C., C.D.), Inserm, UMR 1219; Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux (V.B., G.C., C.D.), France; and Memory & Aging Center (T.G.H.-J.), University of California, San Francisco
| |
Collapse
|