Amyloid and Tau Prediction of Cognitive and Functional Decline in Unimpaired Older Individuals: Longitudinal Data from the A4 and LEARN Studies. Sperling RA, et al, J Prev Alzheimers Dis 2024.

  • Proposé le : 29/09/2024 03:07:32
  • Par : Bot
  • Avec la version du site : v2021_01_12
  • Revu par :
    • Mettre votre nom d'utilisateur
    • Mettre votre nom d'utilisateur
Notes sur les tags :
  • Adopter cette revue :
    Si vous souhaitez prendre en charge cette revue d'article, merci de remplacer le tag Non_attribué par Attribué et ajoutez aussi votre nom d'utilisateur à l'emplacement prévu.
  • Réaliser des modifications :
    Pour modifier ce document, il est nécessaire d'être connecté au site. Pour cela, assurez-vous d'avoir des identifiants valides. Si vous n'en avez pas, contactez-nous. Pour vous connecter, cliquez sur l'icône dans la barre de navigation.
  • Demander la finalisation de la revue de l'article :
    Une fois revue et complétée, merci de remplacer l'étiquette Non_finalisé par A_finaliser. Un administrateur se chargera de valider la revue et de la publier avec le tag Finalisé.

Résumé et points clés

Background: Converging evidence suggests that markers of Alzheimer's disease (AD) pathology in cognitively unimpaired older individuals are associated with high risk of cognitive decline and progression to functional impairment. The Anti-Amyloid Treatment in Asymptomatic Alzheimer's disease (A4) and Longitudinal Evaluation of Amyloid and Neurodegeneration Risk (LEARN) Studies enrolled a large cohort of cognitively normal older individuals across a range of baseline amyloid PET levels. Recent advances in AD blood-based biomarkers further enable the comparison of baseline markers in the prediction of longitudinal clinical outcomes.

Objectives: We sought to evaluate whether biomarker indicators of higher levels of AD pathology at baseline predicted greater cognitive and functional decline, and to compare the relative predictive power of amyloid PET imaging, tau PET imaging, and a plasma P-tau217 assay.

Design: All participants underwent baseline amyloid PET scan, plasma P-tau217; longitudinal cognitive testing with the Primary Alzheimer Cognitive Composite (PACC) every 6 months; and annual functional assessments with the clinical dementia rating (CDR), cognitive functional index (CFI), and activities of daily living (ADL) scales. Baseline tau PET scans were obtained in a subset of participants. Participants with elevated amyloid (Aβ+) on screening PET who met inclusion/exclusion criteria were randomized to receive placebo or solanezumab in a double-blind phase of the A4 Study over 240+ weeks. Participants who did not have elevated amyloid (Aβ-) but were otherwise eligible for the A4 Study were referred to the companion observational LEARN Study with the same outcome assessments over 240+ weeks.

Setting: The A4 and LEARN Studies were conducted at 67 clinical trial sites in the United States, Canada, Japan and Australia.

Participants: Older participants (ages 65-85) who were cognitively unimpaired at baseline (CDR-GS=0, MMSE 25-30 with educational adjustment, and Logical Memory scores within the normal range LMIIa 6-18) were eligible to continue in screening. Aβ+ participants were randomized to either placebo (n=583) or solanezumab (n=564) in the A4 Study. A subset of Aβ+ underwent tau PET imaging in A4 (n=350). Aβ- were enrolled into the LEARN Study (n=553).

Measurements: Baseline 18-F Florbetapir amyloid PET, 18-F Flortaucipir tau PET in a subset and plasma P-tau217 with an electrochemiluminescence (ECL) immunoassay were evaluated as predictors of cognitive (PACC), and functional (CDR, CFI and ADL) change. Models were evaluated to explore the impact of baseline tertiles of amyloid PET and tertiles of plasma P-tau217 on cognitive and functional outcomes in the A4 Study compared to LEARN. Multivariable models were used to evaluate the unique and common variance explained in longitudinal outcomes based on baseline predictors, including effects for age, gender, education, race/ethnic group, APOEε4 carrier status, baseline PACC performance and treatment assignment in A4 participants (solanezumab vs placebo).

Results: Higher baseline amyloid PET CL and P-tau217 levels were associated with faster rates of PACC decline, and increased likelihood of progression to functional impairment (CDR 0.5 or higher on two consecutive measurements), both across LEARN Aβ- and A4 Aβ+ (solanezumab and placebo arms). In analyses considering all baseline predictor variables, P-tau217 was the strongest predictor of PACC decline. Among participants in the highest tertiles of amyloid PET or P-tau217, >50% progressed to CDR 0.5 or greater. In the tau PET substudy, neocortical tau was the strongest predictor of PACC decline, but plasma P-tau217 contributed additional independent predictive variance in commonality variance models.

Conclusions: In a large cohort of cognitively unimpaired individuals enrolled in a Phase 3 clinical trial and companion observational study, these findings confirm that higher baseline levels of amyloid and tau markers are associated with increased rates of cognitive decline and progression to functional impairment. Interestingly, plasma P-tau217 was the best predictor of decline in the overall sample, superior to baseline amyloid PET. Neocortical tau was the strongest predictor of cognitive decline in the subgroup with tau PET, suggesting that tau deposition is most closely linked to clinical decline. These findings indicate that biomarkers of AD pathology are useful to predict decline in an older asymptomatic population and may prove valuable in the selection of individuals for disease-modifying treatments.

Références de l'article

  • Amyloid and Tau Prediction of Cognitive and Functional Decline in Unimpaired Older Individuals: Longitudinal Data from the A4 and LEARN Studies.
  • Amyloid and Tau Prediction of Cognitive and Functional Decline in Unimpaired Older Individuals: Longitudinal Data from the A4 and LEARN Studies.
  • Sperling RA, Donohue MC, Rissman RA, Johnson KA, Rentz DM, Grill JD, Heidebrink JL, Jenkins C, Jimenez-Maggiora G, Langford O, Liu A, Raman R, Yaari R, Holdridge KC, Sims JR, Aisen PS
  • The journal of prevention of Alzheimer's disease
  • 2024
  • J Prev Alzheimers Dis. 2024;11(4):802-813. doi: 10.14283/jpad.2024.122.
  • Humans, *Positron-Emission Tomography, Female, Aged, Male, *tau Proteins/blood, Longitudinal Studies, *Cognitive Dysfunction, *Biomarkers/blood, Alzheimer Disease/blood/diagnostic imaging/drug therapy, Activities of Daily Living, Antibodies, Monoclonal, Humanized/therapeutic use, Amyloid beta-Peptides/blood/metabolism, Aged, 80 and over, Disease Progression, Aniline Compounds
  • Sans_Catégorie, Neurocognitif, Alzheimer, Biomarqueurs
  • Liens
  • Traduction automatique en Français sur Google Translate
  • DOI: 10.14283/jpad.2024.122
  • PMID: 39044488
  • Articles similaires
  • Cité par
  • Références
  • Texte complet gratuit
  • Twitter
  • Twitter cet article (lien vers l'article)
  • Twitter cet article (lien vers cette page)


Discussion

  • Cette section peut être éditée par les relecteurs, les rédacteurs, les modérateurs et les administrateurs. Elle regroupe l'ensemble des échanges autours de la référence ci-dessus présentée.
  • Référez-vous à cette page pour connaître le rôle des utilisateurs et pour participer à la discussion.
  • Il n'y a, pour l'instant, aucune discussion en cours.

Éditer la discussion


Références


Gardez le contact

Suivez notre utilisateur Twitter : @AgingPapers
Nos rencontres visio