Variant-dependent heterogeneity in amyloid β burden in autosomal dominant Alzheimer's disease : cross-sectional and longitudinal analyses of an observational study

Copyright © 2022 Elsevier Ltd. All rights reserved..

BACKGROUND: Insights gained from studying individuals with autosomal dominant Alzheimer's disease have broadly influenced mechanistic hypotheses, biomarker development, and clinical trials in both sporadic and dominantly inherited Alzheimer's disease. Although pathogenic variants causing autosomal dominant Alzheimer's disease are highly penetrant, there is substantial heterogeneity in levels of amyloid β (Aβ) between individuals. We aimed to examine whether this heterogeneity is related to disease progression and to investigate the association with mutation location within PSEN1, PSEN2, or APP.

METHODS: We did cross-sectional and longitudinal analyses of data from the Dominantly Inherited Alzheimer's Network (DIAN) observational study, which enrols individuals from families affected by autosomal dominant Alzheimer's disease. 340 participants in the DIAN study who were aged 18 years or older, had a history of autosomal dominant Alzheimer's disease in their family, and who were enrolled between September, 2008, and June, 2019, were included in our analysis. 206 participants were carriers of pathogenic mutations in PSEN1, PSEN2, or APP, and 134 were non-carriers. 62 unique pathogenic variants were identified in the cohort and were grouped in two ways. First, we sorted variants in PSEN1, PSEN2, or APP by the affected protein domain. Second, we divided PSEN1 variants according to position before or after codon 200. We examined variant-dependent variability in Aβ biomarkers, specifically Pittsburgh-Compound-B PET (PiB-PET) signal, levels of CSF Aβ1-42 (Aβ42), and levels of Aβ1-40 (Aβ40).

FINDINGS: Cortical and striatal PiB-PET signal showed striking variant-dependent variability using both grouping approaches (p<0·0001), despite similar progression on the clinical dementia rating (p>0·7), and CSF Aβ42 levels (codon-based grouping: p=0·49; domain-based grouping: p=0·095). Longitudinal PiB-PET signal also varied across codon-based groups, mirroring cross-sectional analyses.

INTERPRETATION: Autosomal dominant Alzheimer's disease pathogenic variants showed highly differential temporal and regional patterns of PiB-PET signal, despite similar functional progression. These findings suggest that although increased PiB-PET signal is generally seen in autosomal dominant Alzheimer's disease, higher levels of PiB-PET signal at an individual level might not reflect more severe or more advanced disease. Our results have high relevance for ongoing clinical trials in autosomal dominant Alzheimer's disease, including those using Aβ PET as a surrogate marker of disease progression. Additionally, and pertinent to both sporadic and autosomal dominant Alzheimer's disease, our results suggest that CSF and PET measures of Aβ levels are not interchangeable and might reflect different Aβ-driven pathobiological processes.

FUNDING: National Institute on Aging, Doris Duke Charitable Foundation, German Center for Neurodegenerative Diseases, Japanese Agency for Medical Research and Development.

Errataetall:

CommentIn: Lancet Neurol. 2022 Feb;21(2):105-106. - PMID 35065027

Medienart:

E-Artikel

Erscheinungsjahr:

2022

Erschienen:

2022

Enthalten in:

Zur Gesamtaufnahme - volume:21

Enthalten in:

The Lancet. Neurology - 21(2022), 2 vom: 29. Feb., Seite 140-152

Sprache:

Englisch

Beteiligte Personen:

Chhatwal, Jasmeer P [VerfasserIn]
Schultz, Stephanie A [VerfasserIn]
McDade, Eric [VerfasserIn]
Schultz, Aaron P [VerfasserIn]
Liu, Lei [VerfasserIn]
Hanseeuw, Bernard J [VerfasserIn]
Joseph-Mathurin, Nelly [VerfasserIn]
Feldman, Rebecca [VerfasserIn]
Fitzpatrick, Colleen D [VerfasserIn]
Sparks, Kathryn P [VerfasserIn]
Levin, Johannes [VerfasserIn]
Berman, Sarah B [VerfasserIn]
Renton, Alan E [VerfasserIn]
Esposito, Bianca T [VerfasserIn]
Fernandez, Maria Vitoria [VerfasserIn]
Sung, Yun Ju [VerfasserIn]
Lee, Jae Hong [VerfasserIn]
Klunk, William E [VerfasserIn]
Hofmann, Anna [VerfasserIn]
Noble, James M [VerfasserIn]
Graff-Radford, Neill [VerfasserIn]
Mori, Hiroshi [VerfasserIn]
Salloway, Steven M [VerfasserIn]
Masters, Colin L [VerfasserIn]
Martins, Ralph [VerfasserIn]
Karch, Celeste M [VerfasserIn]
Xiong, Chengjie [VerfasserIn]
Cruchaga, Carlos [VerfasserIn]
Perrin, Richard J [VerfasserIn]
Gordon, Brian A [VerfasserIn]
Benzinger, Tammie L S [VerfasserIn]
Fox, Nick C [VerfasserIn]
Schofield, Peter R [VerfasserIn]
Fagan, Anne M [VerfasserIn]
Goate, Alison M [VerfasserIn]
Morris, John C [VerfasserIn]
Bateman, Randall J [VerfasserIn]
Johnson, Keith A [VerfasserIn]
Sperling, Reisa A [VerfasserIn]
Dominantly Inherited Alzheimer's Network Investigators [VerfasserIn]

Links:

Volltext

Themen:

Amyloid beta-Peptides
Biomarkers
Journal Article
Observational Study
Research Support, N.I.H., Extramural
Research Support, Non-U.S. Gov't

Anmerkungen:

Date Completed 21.03.2022

Date Revised 01.06.2023

published: Print

CommentIn: Lancet Neurol. 2022 Feb;21(2):105-106. - PMID 35065027

Citation Status MEDLINE

doi:

10.1016/S1474-4422(21)00375-6

funding:

Förderinstitution / Projekttitel:

PPN (Katalog-ID):

NLM335996124