51
|
Guo X, Chen K, Chen Y, Xiong C, Su Y, Yao L, Reiman EM. A Computational Monte Carlo Simulation Strategy to Determine the Temporal Ordering of Abnormal Age Onset Among Biomarkers of Alzheimer's Disease. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2613-2622. [PMID: 34428151 PMCID: PMC9588284 DOI: 10.1109/tcbb.2021.3106939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
To quantitatively determining the temporal ordering of abnormal age onsets (AAO) among various biomarkers for Alzheimer's disease (AD), we introduced a computational Monte-Carlo simulation (CMCS) to statistically examine such ordering of an AAO pair or over all AAOs. The CMCS 1) simulates longitudinal data, estimates AAO for each iteration, and finally assesses the type-I error of an AAO pair or all AAO ordering. Using hippocampus volume (VHC), cerebral glucose hypometabolic convergence index (HCI), plasma neurofilament light (NfL), mini-mental state exam (MMSE), the auditory verbal learning test-long term memory (AVLT-LTM), short term memory (AVLT-STM) and clinical-dementia rating sum of box scale (CDR-SOB) from 382 mild cognitive impairment converters and non-converters, the CMCS estimated type-I error for the earlier AAO of VHC, AVLT_STM and AVLT_LTM each than MMSE was significant (p<0.002). The type-I error for the overall AAO temporal ordering of VHC ≤ AVLT_STM ≤ AVLT_LTM < HCI ≤ MMSE ≤ CDR-SOB ≤ NfL was p = 0.012. These findings showed that our CMCS is capable of providing statistical inferences for quantifying AAO ordering which has important implications in advancing our understanding of AD.
Collapse
|
52
|
Puig-Pijoan A, García-Escobar G, Fernández-Lebrero A, Manero Borràs R, Sánchez-Benavides G, Navalpotro-Gómez I, Cascales Lahoz D, Suárez-Calvet M, Grau-Rivera O, Boltes Alandí A, Pont-Sunyer M, Ortiz-Gil J, Carrillo-Molina S, López-Villegas D, Abellán-Vidal M, Martínez-Casamitjana M, Hernández-Sánchez J, Peña-Casanova J, Roquer J, Padrós Fluvià A, Puente-Périz V. Estudio CORCOBIA: determinación de puntos de corte de biomarcadores de enfermedad de Alzheimer en LCR en una cohorte clínica. Neurologia 2022. [DOI: 10.1016/j.nrl.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
53
|
Ebenau JL, Pelkmans W, Verberk IMW, Verfaillie SCJ, van den Bosch KA, van Leeuwenstijn M, Collij LE, Scheltens P, Prins ND, Barkhof F, van Berckel BNM, Teunissen CE, van der Flier WM. Association of CSF, Plasma, and Imaging Markers of Neurodegeneration With Clinical Progression in People With Subjective Cognitive Decline. Neurology 2022; 98:e1315-e1326. [PMID: 35110378 PMCID: PMC8967429 DOI: 10.1212/wnl.0000000000200035] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Multiple biomarkers have been suggested to measure neurodegeneration (N) in the AT(N) framework, leading to inconsistencies between studies. We investigated the association of 5 N biomarkers with clinical progression and cognitive decline in individuals with subjective cognitive decline (SCD). METHODS We included individuals with SCD from the Amsterdam Dementia Cohort and SCIENCe project, a longitudinal cohort study (follow-up 4±3 years). We used the following N biomarkers: CSF total tau (t-tau), medial temporal atrophy visual rating on MRI, hippocampal volume (HV), serum neurofilament light (NfL), and serum glial fibrillary acidic protein (GFAP). We determined correlations between biomarkers. We assessed associations between N biomarkers and clinical progression to mild cognitive impairment or dementia (Cox regression) and Mini-Mental State Examination (MMSE) over time (linear mixed models). Models included age, sex, CSF β-amyloid (Aβ) (A), and CSF p-tau (T) as covariates, in addition to the N biomarker. RESULT We included 401 individuals (61±9 years, 42% female, MMSE 28 ± 2, vascular comorbidities 8%-19%). N biomarkers were modestly to moderately correlated (range r -0.28 - 0.58). Serum NfL and GFAP correlated most strongly (r 0.58, p < 0.01). T-tau was strongly correlated with p-tau (r 0.89, p < 0.01), although these biomarkers supposedly represent separate biomarker groups. All N biomarkers individually predicted clinical progression, but only HV, NfL, and GFAP added predictive value beyond Aβ and p-tau (hazard ratio 1.52 [95% CI 1.11-2.09]; 1.51 [1.05-2.17]; 1.50 [1.04-2.15]). T-tau, HV, and GFAP individually predicted MMSE slope (range β -0.17 to -0.11, p < 0.05), but only HV remained associated beyond Aβ and p-tau (β -0.13 [SE 0.04]; p < 0.05). DISCUSSION In cognitively unimpaired older adults, correlations between different N biomarkers were only moderate, indicating they reflect different aspects of neurodegeneration and should not be used interchangeably. T-tau was strongly associated with p-tau (T), which makes it less desirable to use as a measure for N. HV, NfL, and GFAP predicted clinical progression beyond A and T. Our results do not allow to choose one most suitable biomarker for N, but illustrate the added prognostic value of N beyond A and T. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that HV, NfL, and GFAP predicted clinical progression beyond A and T in individuals with SCD.
Collapse
Affiliation(s)
- Jarith L Ebenau
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK.
| | - Wiesje Pelkmans
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Inge M W Verberk
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Sander C J Verfaillie
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Karlijn A van den Bosch
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Mardou van Leeuwenstijn
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Lyduine E Collij
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Philip Scheltens
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Niels D Prins
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Frederik Barkhof
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Bart N M van Berckel
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Charlotte E Teunissen
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Wiesje M van der Flier
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| |
Collapse
|
54
|
Huang S, Wang YJ, Guo J. Biofluid Biomarkers of Alzheimer’s Disease: Progress, Problems, and Perspectives. Neurosci Bull 2022; 38:677-691. [PMID: 35306613 PMCID: PMC9206048 DOI: 10.1007/s12264-022-00836-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Abstract
Since the establishment of the biomarker-based A-T-N (Amyloid/Tau/Neurodegeneration) framework in Alzheimer’s disease (AD), the diagnosis of AD has become more precise, and cerebrospinal fluid tests and positron emission tomography examinations based on this framework have become widely accepted. However, the A-T-N framework does not encompass the whole spectrum of AD pathologies, and problems with invasiveness and high cost limit the application of the above diagnostic methods aimed at the central nervous system. Therefore, we suggest the addition of an “X” to the A-T-N framework and a focus on peripheral biomarkers in the diagnosis of AD. In this review, we retrospectively describe the recent progress in biomarkers based on the A-T-N-X framework, analyze the problems, and present our perspectives on the diagnosis of AD.
Collapse
|
55
|
Liu Y, Han PR, Hu H, Wang ZT, Guo Y, Ou YN, Cao XP, Tan L, Yu JT. A Multi-Dimensional Comparison of Alzheimer's Disease Neurodegenerative Biomarkers. J Alzheimers Dis 2022; 87:197-209. [PMID: 35275546 DOI: 10.3233/jad-215724] [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: 11/15/2022]
Abstract
BACKGROUND In the 2018 AT(N) framework, neurodegenerative (N) biomarkers plays an essential role in the research and staging of Alzheimer's disease (AD); however, the different choice of N may result in discordances. OBJECTIVE We aimed to compare different potential N biomarkers. METHODS We examined these N biomarkers among 1,238 participants from Alzheimer's Disease Neuroimaging Initiative (ADNI) in their 1) diagnostic utility, 2) cross-sectional and longitudinal correlations between different N biomarkers and clinical variables, and 3) the conversion risk of different N profiles. RESULTS Six neurodegenerative biomarkers changed significantly from preclinical AD, through prodromal AD to AD dementia stage, thus they were chosen as the candidate N biomarkers: hippocampal volume (HV), 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET), cerebrospinal fluid (CSF), total tau (T-tau), plasma neurofilament light chain (NFL), CSF NFL, and CSF neurogranin (Ng). Results indicated that FDG-PET not only had the greatest diagnostic utility in differentiating AD from controls (area under the curve: FDG-PET, 0.922), but also had the strongest association with cognitive scores. Furthermore, FDG-PET positive group showed the fastest memory decline (hazard ratio: FDG-PET, 3.45), which was also true even in the presence of amyloid-β pathology. Moreover, we observed great discordances between three valuable N biomarkers (FDG-PET, HV, and T-tau). CONCLUSION These results underline the importance of using FDG-PET as N in terms of cognitive decline and AD conversion, followed by HV, and could be a great complement to the AT(N) framework.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Pei-Ran Han
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | | |
Collapse
|
56
|
Karran E, De Strooper B. The amyloid hypothesis in Alzheimer disease: new insights from new therapeutics. Nat Rev Drug Discov 2022; 21:306-318. [PMID: 35177833 DOI: 10.1038/s41573-022-00391-w] [Citation(s) in RCA: 368] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 12/14/2022]
Abstract
Many drugs that target amyloid-β (Aβ) in Alzheimer disease (AD) have failed to demonstrate clinical efficacy. However, four anti-Aβ antibodies have been shown to mediate the removal of amyloid plaque from brains of patients with AD, and the FDA has recently granted accelerated approval to one of these, aducanumab, using reduction of amyloid plaque as a surrogate end point. The rationale for approval and the extent of the clinical benefit from these antibodies are under intense debate. With the aim of informing this debate, we review clinical trial data for drugs that target Aβ from the perspective of the temporal interplay between the two pathognomonic protein aggregates in AD - Aβ plaques and tau neurofibrillary tangles - and their relationship to cognitive impairment, highlighting differences in drug properties that could affect their clinical performance. On this basis, we propose that Aβ pathology drives tau pathology, that amyloid plaque would need to be reduced to a low level (~20 centiloids) to reveal significant clinical benefit and that there will be a lag between the removal of amyloid and the potential to observe a clinical benefit. We conclude that the speed of amyloid removal from the brain by a potential therapy will be important in demonstrating clinical benefit in the context of a clinical trial.
Collapse
Affiliation(s)
- Eric Karran
- Cambridge Research Center, AbbVie, Inc., Cambridge, MA, USA.
| | - Bart De Strooper
- VIB Centre for Brain Disease Research, KU Leuven, Leuven, Belgium.,UK Dementia Research Institute, University College London, London, UK
| |
Collapse
|
57
|
Janelidze S, Palmqvist S, Leuzy A, Stomrud E, Verberk IMW, Zetterberg H, Ashton NJ, Pesini P, Sarasa L, Allué JA, Teunissen CE, Dage JL, Blennow K, Mattsson-Carlgren N, Hansson O. Detecting amyloid positivity in early Alzheimer's disease using combinations of plasma Aβ42/Aβ40 and p-tau. Alzheimers Dement 2022; 18:283-293. [PMID: 34151519 DOI: 10.1002/alz.12395] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 01/20/2023]
Abstract
INTRODUCTION We studied usefulness of combining blood amyloid beta (Aβ)42/Aβ40, phosphorylated tau (p-tau)217, and neurofilament light (NfL) to detect abnormal brain Aβ deposition in different stages of early Alzheimer's disease (AD). METHODS Plasma biomarkers were measured using mass spectrometry (Aβ42/Aβ40) and immunoassays (p-tau217 and NfL) in cognitively unimpaired individuals (CU, N = 591) and patients with mild cognitive impairment (MCI, N = 304) from two independent cohorts (BioFINDER-1, BioFINDER-2). RESULTS In CU, a combination of plasma Aβ42/Aβ40 and p-tau217 detected abnormal brain Aβ status with area under the curve (AUC) of 0.83 to 0.86. In MCI, the models including p-tau217 alone or Aβ42/Aβ40 and p-tau217 had similar AUCs (0.86-0.88); however, the latter showed improved model fit. The models were implemented in an online application providing individualized risk assessments (https://brainapps.shinyapps.io/PredictABplasma/). DISCUSSION A combination of plasma Aβ42/Aβ40 and p-tau217 discriminated Aβ status with relatively high accuracy, whereas p-tau217 showed strongest associations with Aβ pathology in MCI but not in CU.
Collapse
Affiliation(s)
- Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Skåne University Hospital, Malmö, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Skåne University Hospital, Malmö, Sweden
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - 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, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute 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
| | | | | | | | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Kaj Blennow
- 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
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
58
|
Sacchi L, Carandini T, Fumagalli GG, Pietroboni AM, Contarino VE, Siggillino S, Arcaro M, Fenoglio C, Zito F, Marotta G, Castellani M, Triulzi F, Galimberti D, Scarpini E, Arighi A. Unravelling the Association Between Amyloid-PET and Cerebrospinal Fluid Biomarkers in the Alzheimer's Disease Spectrum: Who Really Deserves an A+? J Alzheimers Dis 2021; 85:1009-1020. [PMID: 34897084 DOI: 10.3233/jad-210593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Association between cerebrospinal fluid (CSF)-amyloid-β (Aβ)42 and amyloid-PET measures is inconstant across the Alzheimer's disease (AD) spectrum. However, they are considered interchangeable, along with Aβ 42/40 ratio, for defining 'Alzheimer's Disease pathologic change' (A+). OBJECTIVE Herein, we further characterized the association between amyloid-PET and CSF biomarkers and tested their agreement in a cohort of AD spectrum patients. METHODS We include ed 23 patients who underwent amyloid-PET, MRI, and CSF analysis showing reduced levels of Aβ 42 within a 365-days interval. Thresholds used for dichotomization were: Aβ 42 < 640 pg/mL (Aβ 42+); pTau > 61 pg/mL (pTau+); and Aβ 42/40 < 0.069 (ADratio+). Amyloid-PET scans were visually assessed and processed by four pipelines (SPMCL, SPMAAL, FSGM, FSWC). RESULTS Different pipelines gave highly inter-correlated standardized uptake value ratios (SUVRs) (rho = 0.93-0.99). The most significant findings were: pTau positive correlation with SPMCL SUVR (rho = 0.56, p = 0.0063) and Aβ 42/40 negative correlation with SPMCL and SPMAAL SUVRs (rho = -0.56, p = 0.0058; rho = -0.52, p = 0.0117 respectively). No correlations between CSF-Aβ 42 and global SUVRs were observed. In subregion analysis, both pTau and Aβ 42/40 values significantly correlated with cingulate SUVRs from any pipeline (R2 = 0.55-0.59, p < 0.0083), with the strongest associations observed for the posterior/isthmus cingulate areas. However, only associations observed for Aβ 42/40 ratio were still significant in linear regression models. Moreover, combining pTau with Aβ 42 or using Aβ 42/40, instead of Aβ 42 alone, increased concordance with amyloid-PET status from 74% to 91% based on visual reads and from 78% to 96% based on Centiloids. CONCLUSION We confirmed that, in the AD spectrum, amyloid-PET measures show a stronger association and a better agreement with CSF-Aβ 42/40 and secondarily pTau rather than Aβ 42 levels.
Collapse
Affiliation(s)
- Luca Sacchi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Anna Margherita Pietroboni
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Silvia Siggillino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marina Arcaro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Felicia Zito
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Marotta
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Castellani
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elio Scarpini
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Arighi
- University of Milan, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
59
|
Kühnel L, Bouteloup V, Lespinasse J, Chêne G, Dufouil C, Molinuevo JL, Raket LL. Personalized prediction of progression in pre-dementia patients based on individual biomarker profile: A development and validation study. Alzheimers Dement 2021; 17:1938-1949. [PMID: 34581496 DOI: 10.1002/alz.12363] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 11/10/2022]
Abstract
INTRODUCTION The prognosis of patients at the pre-dementia stage is difficult to define. The aim of this study is to develop and validate a biomarker-based continuous model for predicting the individual cognitive level at any future moment. In addition to personalized prognosis, such a model could reduce trial sample size requirements by allowing inclusion of a homogenous patient population. METHODS Disease-progression modeling of longitudinal cognitive scores of pre-dementia patients (baseline Clinical Dementia Rating ≤ 0.5) was used to derive a biomarker profile that was predictive of patient's cognitive progression along the dementia continuum. The biomarker profile model was developed and validated in the MEMENTO cohort and externally validated in the Alzheimer's Disease Neuroimaging Initiative. RESULTS Of nine candidate biomarkers in the development analysis, three cerebrospinal fluid and two magnetic resonance imaging measures were selected to form the final biomarker profile. The model-based prognosis of individual future cognitive deficit was shown to significantly improve when incorporating biomarker information on top of cognition and demographic data. In trial power calculations, adjusting the primary analysis for the baseline biomarker profile reduced sample size requirements by ≈10%. Compared to conventional cognitive cut-offs, inclusion criteria based on biomarker-profile cut-offs resulted in up to 28% reduced sample size requirements due to increased homogeneity in progression patterns. DISCUSSION The biomarker profile allows prediction of personalized trajectories of future cognitive progression. This enables accurate personalized prognosis in clinical care and better selection of patient populations for clinical trials. A web-based application for prediction of patients' future cognitive progression is available online.
Collapse
Affiliation(s)
- Line Kühnel
- H. Lundbeck A/S, Copenhagen, Denmark.,Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vincent Bouteloup
- Inserm, Population Health Research Center, University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Pole Santé Publique, Talence, France
| | - Jérémie Lespinasse
- Inserm, Population Health Research Center, University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Pole Santé Publique, Talence, France
| | - Geneviève Chêne
- Inserm, Population Health Research Center, University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Pole Santé Publique, Talence, France
| | - Carole Dufouil
- Inserm, Population Health Research Center, University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Pole Santé Publique, Talence, France
| | | | - Lars Lau Raket
- H. Lundbeck A/S, Copenhagen, Denmark.,Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | |
Collapse
|
60
|
Hassenstab J, Nicosia J, LaRose M, Aschenbrenner AJ, Gordon BA, Benzinger TLS, Xiong C, Morris JC. Is comprehensiveness critical? Comparing short and long format cognitive assessments in preclinical Alzheimer disease. Alzheimers Res Ther 2021; 13:153. [PMID: 34517889 PMCID: PMC8436865 DOI: 10.1186/s13195-021-00894-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/24/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Comprehensive testing of cognitive functioning is standard practice in studies of Alzheimer disease (AD). Short-form tests like the Montreal Cognitive Assessment (MoCA) use a "sampling" of measures, administering key items in a shortened format to efficiently assess cognition while reducing time requirements, participant burden, and administrative costs. We compared the MoCA to a commonly used long-form cognitive battery in predicting AD symptom onset and sensitivity to AD neuroimaging biomarkers. METHODS Survival, area under the receiver operating characteristic (ROC) curve (AUC), and multiple regression analyses compared the MoCA and long-form measures in predicting time to symptom onset in cognitively normal older adults (n = 6230) from the National Alzheimer's Coordinating Center (NACC) cohort who had, on average, 2.3 ± 1.2 annual assessments. Multiple regression models in a separate sample (n = 416) from the Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC) compared the sensitivity of the MoCA and long-form measures to neuroimaging biomarkers including amyloid PET, tau PET, and cortical thickness. RESULTS Hazard ratios suggested that both the MoCA and the long-form measures are similarly and modestly efficacious in predicting symptomatic conversion, although model comparison analyses indicated that the long-form measures slightly outperformed the MoCA (HRs > 1.57). AUC analyses indicated no difference between the measures in predicting conversion (DeLong's test, Z = 1.48, p = 0.13). Sensitivity to AD neuroimaging biomarkers was similar for the two measures though there were only modest associations with tau PET (rs = - 0.13, ps < 0.02) and cortical thickness in cognitively normal participants (rs = 0.15-0.16, ps < 0.007). CONCLUSIONS Both test formats showed weak associations with symptom onset, AUC analyses indicated low diagnostic accuracy, and biomarker correlations were modest in cognitively normal participants. Alternative assessment approaches are needed to improve how clinicians and researchers monitor cognitive changes and disease progression prior to symptom onset.
Collapse
Affiliation(s)
- Jason Hassenstab
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA.
| | - Jessica Nicosia
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan LaRose
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
61
|
Developing the ATX(N) classification for use across the Alzheimer disease continuum. Nat Rev Neurol 2021; 17:580-589. [PMID: 34239130 DOI: 10.1038/s41582-021-00520-w] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Breakthroughs in the development of highly accurate fluid and neuroimaging biomarkers have catalysed the conceptual transformation of Alzheimer disease (AD) from the traditional clinical symptom-based definition to a clinical-biological construct along a temporal continuum. The AT(N) system is a symptom-agnostic classification scheme that categorizes individuals using biomarkers that chart core AD pathophysiological features, namely the amyloid-β (Aβ) pathway (A), tau-mediated pathophysiology (T) and neurodegeneration (N). This biomarker matrix is now expanding towards an ATX(N) system, where X represents novel candidate biomarkers for additional pathophysiological mechanisms such as neuroimmune dysregulation, synaptic dysfunction and blood-brain barrier alterations. In this Perspective, we describe the conceptual framework and clinical importance of the existing AT(N) system and the evolving ATX(N) system. We provide a state-of-the-art summary of the potential contexts of use of these systems in AD clinical trials and future clinical practice. We also discuss current challenges related to the validation, standardization and qualification process and provide an outlook on the real-world application of the AT(N) system.
Collapse
|
62
|
Yu M, Sporns O, Saykin AJ. The human connectome in Alzheimer disease - relationship to biomarkers and genetics. Nat Rev Neurol 2021; 17:545-563. [PMID: 34285392 PMCID: PMC8403643 DOI: 10.1038/s41582-021-00529-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The pathology of Alzheimer disease (AD) damages structural and functional brain networks, resulting in cognitive impairment. The results of recent connectomics studies have now linked changes in structural and functional network organization in AD to the patterns of amyloid-β and tau accumulation and spread, providing insights into the neurobiological mechanisms of the disease. In addition, the detection of gene-related connectome changes might aid in the early diagnosis of AD and facilitate the development of personalized therapeutic strategies that are effective at earlier stages of the disease spectrum. In this article, we review studies of the associations between connectome changes and amyloid-β and tau pathologies as well as molecular genetics in different subtypes and stages of AD. We also highlight the utility of connectome-derived computational models for replicating empirical findings and for tracking and predicting the progression of biomarker-indicated AD pathophysiology.
Collapse
Affiliation(s)
- Meichen Yu
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
| | - Olaf Sporns
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Network Science Institute, Bloomington, IN, USA.
| |
Collapse
|
63
|
Gouilly D, Tisserand C, Nogueira L, Saint-Lary L, Rousseau V, Benaiteau M, Rafiq M, Carlier J, Milongo-Rigal E, Pagès JC, Pariente J. Taking the A Train? Limited Consistency of Aβ42 and the Aβ42/40 Ratio in the AT(N) Classification. J Alzheimers Dis 2021; 83:1033-1038. [PMID: 34397413 DOI: 10.3233/jad-210236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The consistency of cerebrospinal fluid amyloid-β (Aβ)42/40 ratio and Aβ 42 has not been assessed in the AT(N) classification system. We analyzed the classification changes of the dichotomized amyloid status (A+/A-) in 363 patients tested for Alzheimer's disease biomarkers after Aβ 42 was superseded by the Aβ 42/40 ratio. The consistency of Aβ 42 and the Aβ 42/40 ratio was very low. Notably, the proportions of "false" A+T-patients were considerable (74-91%) and corresponded mostly to patients not clinically diagnosed with Alzheimer's disease. Our results suggest that the interchangeability of Aβ 42/40 ratio and Aβ 42 is limited for classifying patients in clinical setting using the AT(N) scheme.
Collapse
Affiliation(s)
| | - Camille Tisserand
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Leonor Nogueira
- Department of Cell Biology and Cytology, CHU Toulouse Purpan, France
| | - Laura Saint-Lary
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - Vanessa Rousseau
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - Marie Benaiteau
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Marie Rafiq
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Jasmine Carlier
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - Emilie Milongo-Rigal
- Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | | | - Jérémie Pariente
- Toulouse Neuroimaging Center, Toulouse, France.,Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| |
Collapse
|
64
|
Lin RR, Xue YY, Li XY, Chen YH, Tao QQ, Wu ZY. Optimal Combinations of AT(N) Biomarkers to Determine Longitudinal Cognition in the Alzheimer's Disease. Front Aging Neurosci 2021; 13:718959. [PMID: 34421579 PMCID: PMC8377373 DOI: 10.3389/fnagi.2021.718959] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/05/2021] [Indexed: 01/12/2023] Open
Abstract
Background: National Institute on Aging-Alzheimer's Association (NIA-AA) proposed the AT(N) system based on β-amyloid deposition, pathologic tau, and neurodegeneration, which considered the definition of Alzheimer's disease (AD) as a biological construct. However, the associations between different AT(N) combinations and cognitive progression have been poorly explored systematically. The aim of this study is to compare different AT(N) combinations using recognized biomarkers within the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Methods: A total of 341 participants were classified into cognitively unimpaired (CU; n = 200) and cognitively impaired (CI; n = 141) groups according to the clinical manifestations and neuropsychological tests. Cerebrospinal fluid (CSF) Aβ42 and amyloid-PET ([18F]flutemetamol) were used as biomarkers for A; CSF phosphorylated tau (p-tau) and tau-PET ([18F]flortaucipir) were used as biomarkers for T; CSF total tau (t-tau), hippocampal volume, temporal cortical thickness, [18F]fluorodeoxyglucose (FDG) PET, and plasma neurofilament light (NfL) were used as biomarkers for (N). Binary biomarkers were obtained from the Youden index and publicly available cutoffs. Prevalence of AT(N) categories was compared between different biomarkers within the group using related independent sample non-parametric test. The relationship between AT(N) combinations and 12-year longitudinal cognition was assessed using linear mixed-effects modeling. Results: Among the CU participants, A-T-(N)- was most common. More T+ were detected using p-tau than tau PET (p < 0.05), and more (N)+ were observed using fluid biomarkers (p < 0.001). A+T+(N)+ was more common in the CI group. Tau PET combined with cortical thickness best predicted cognitive changes in the CI group and MRI predicted changes in the CU group. Conclusions: These findings suggest that optimal AT(N) combinations to determine longitudinal cognition differ by cognitive status. Different biomarkers within a specific component for defining AT(N) cannot be used identically. Furthermore, different strategies for discontinuous biomarkers will be an important area for future studies.
Collapse
Affiliation(s)
| | | | | | | | - Qing-Qing Tao
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
65
|
Ossenkoppele R, Reimand J, Smith R, Leuzy A, Strandberg O, Palmqvist S, Stomrud E, Zetterberg H, Scheltens P, Dage JL, Bouwman F, Blennow K, Mattsson-Carlgren N, Janelidze S, Hansson O. Tau PET correlates with different Alzheimer's disease-related features compared to CSF and plasma p-tau biomarkers. EMBO Mol Med 2021; 13:e14398. [PMID: 34254442 PMCID: PMC8350902 DOI: 10.15252/emmm.202114398] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
PET, CSF and plasma biomarkers of tau pathology may be differentially associated with Alzheimer's disease (AD)‐related demographic, cognitive, genetic and neuroimaging markers. We examined 771 participants with normal cognition, mild cognitive impairment or dementia from BioFINDER‐2 (n = 400) and ADNI (n = 371). All had tau‐PET ([18F]RO948 in BioFINDER‐2, [18F]flortaucipir in ADNI) and CSF p‐tau181 biomarkers available. Plasma p‐tau181 and plasma/CSF p‐tau217 were available in BioFINDER‐2 only. Concordance between PET, CSF and plasma tau biomarkers ranged between 66 and 95%. Across the whole group, ridge regression models showed that increased CSF and plasma p‐tau181 and p‐tau217 levels were independently of tau PET associated with higher age, and APOEɛ4‐carriership and Aβ‐positivity, while increased tau‐PET signal in the temporal cortex was associated with worse cognitive performance and reduced cortical thickness. We conclude that biofluid and neuroimaging markers of tau pathology convey partly independent information, with CSF and plasma p‐tau181 and p‐tau217 levels being more tightly linked with early markers of AD (especially Aβ‐pathology), while tau‐PET shows the strongest associations with cognitive and neurodegenerative markers of disease progression.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Juhan Reimand
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Health Technologies, Tallinn University of Technology, Tallinn, Estonia.,Radiology Centre, North Estonia Medical Centre, Tallinn, Estonia
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Femke Bouwman
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | | | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
66
|
Ingala S, De Boer C, Masselink LA, Vergari I, Lorenzini L, Blennow K, Chételat G, Di Perri C, Ewers M, van der Flier WM, Fox NC, Gispert JD, Haller S, Molinuevo JL, Muniz‐Terrera G, Mutsaerts HJMM, Ritchie CW, Ritchie K, Schmidt M, Schwarz AJ, Vermunt L, Waldman AD, Wardlaw J, Wink AM, Wolz R, Wottschel V, Scheltens P, Visser PJ, Barkhof F. Application of the ATN classification scheme in a population without dementia: Findings from the EPAD cohort. Alzheimers Dement 2021; 17:1189-1204. [PMID: 33811742 PMCID: PMC8359976 DOI: 10.1002/alz.12292] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/11/2020] [Accepted: 12/22/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND We classified non-demented European Prevention of Alzheimer's Dementia (EPAD) participants through the amyloid/tau/neurodegeneration (ATN) scheme and assessed their neuropsychological and imaging profiles. MATERIALS AND METHODS From 1500 EPAD participants, 312 were excluded. Cerebrospinal fluid cut-offs of 1000 pg/mL for amyloid beta (Aß)1-42 and 27 pg/mL for p-tau181 were validated using Gaussian mixture models. Given strong correlation of p-tau and t-tau (R2 = 0.98, P < 0.001), neurodegeneration was defined by age-adjusted hippocampal volume. Multinomial regressions were used to test whether neuropsychological tests and regional brain volumes could distinguish ATN stages. RESULTS Age was 65 ± 7 years, with 58% females and 38% apolipoprotein E (APOE) ε4 carriers; 57.1% were A-T-N-, 32.5% were in the Alzheimer's disease (AD) continuum, and 10.4% suspected non-Alzheimer's pathology. Age and cerebrovascular burden progressed with biomarker positivity (P < 0.001). Cognitive dysfunction appeared with T+. Paradoxically higher regional gray matter volumes were observed in A+T-N- compared to A-T-N- (P < 0.001). DISCUSSION In non-demented individuals along the AD continuum, p-tau drives cognitive dysfunction. Memory and language domains are affected in the earliest stages.
Collapse
Affiliation(s)
- Silvia Ingala
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Casper De Boer
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Larissa A Masselink
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Ilaria Vergari
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Luigi Lorenzini
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain @ Caen‐NormandieCyceronCaenFrance
| | - Carol Di Perri
- Centre for Dementia PreventionEdinburgh Imaging, UK Dementia Research Institute at The University of EdinburghEdinburghUK
| | - Michael Ewers
- Institute for Stroke and Dementia ResearchKlinikum der Universitat MünchenLudwig‐Maximilians‐Universitat LMUMunichGermany
| | - Wiesje M van der Flier
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Nick C Fox
- Dementia Research CentreDepartment of Neurodegenerative Disease & UK Dementia Research InstituteInstitute of NeurologyUniversity College LondonLondonUK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Sven Haller
- CIRD Centre d'Imagerie Rive DroiteGenevaSwitzerland
| | - José Luís Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hopsital Clínic‐IDIBAPSAlzheimer's Disease & Other Cognitive Disorders UnitBarcelonaSpain
| | - Graciela Muniz‐Terrera
- Centre for Dementia PreventionEdinburgh Imaging, UK Dementia Research Institute at The University of EdinburghEdinburghUK
| | - Henri JMM Mutsaerts
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
- Ghent Institute for Functional and Metabolic Imaging (GIfMI)Ghent UniversityGhentBelgium
| | - Craig W Ritchie
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Karen Ritchie
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | | | - Adam J Schwarz
- Takeda Pharmaceutical Company LtdCambridgeMassachusettsUSA
| | - Lisa Vermunt
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Adam D Waldman
- Centre for Dementia PreventionEdinburgh Imaging, UK Dementia Research Institute at The University of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Joanna Wardlaw
- Centre for Dementia PreventionEdinburgh Imaging, UK Dementia Research Institute at The University of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Alle Meije Wink
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | | | - Viktor Wottschel
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Philip Scheltens
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
- Department of Psychiatry & NeuropsychologySchool for Mental Health and NeuroscienceMaastricht UniversityMaastrichtthe Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
- Institutes of Neurology and Healthcare EngineeringUniversity College LondonLondonUK
| | - the EPAD consortium
- Department of Radiology and Nuclear MedicineAmsterdam UMC Location VUmcVrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamthe Netherlands
| |
Collapse
|
67
|
Palmqvist S, Tideman P, Cullen N, Zetterberg H, Blennow K, Dage JL, Stomrud E, Janelidze S, Mattsson-Carlgren N, Hansson O. Prediction of future Alzheimer's disease dementia using plasma phospho-tau combined with other accessible measures. Nat Med 2021; 27:1034-1042. [PMID: 34031605 DOI: 10.1038/s41591-021-01348-z] [Citation(s) in RCA: 276] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/12/2021] [Indexed: 02/04/2023]
Abstract
A combination of plasma phospho-tau (P-tau) and other accessible biomarkers might provide accurate prediction about the risk of developing Alzheimer's disease (AD) dementia. We examined this in participants with subjective cognitive decline and mild cognitive impairment from the BioFINDER (n = 340) and Alzheimer's Disease Neuroimaging Initiative (ADNI) (n = 543) studies. Plasma P-tau, plasma Aβ42/Aβ40, plasma neurofilament light, APOE genotype, brief cognitive tests and an AD-specific magnetic resonance imaging measure were examined using progression to AD as outcome. Within 4 years, plasma P-tau217 predicted AD accurately (area under the curve (AUC) = 0.83) in BioFINDER. Combining plasma P-tau217, memory, executive function and APOE produced higher accuracy (AUC = 0.91, P < 0.001). In ADNI, this model had similar AUC (0.90) using plasma P-tau181 instead of P-tau217. The model was implemented online for prediction of the individual probability of progressing to AD. Within 2 and 6 years, similar models had AUCs of 0.90-0.91 in both cohorts. Using cerebrospinal fluid P-tau, Aβ42/Aβ40 and neurofilament light instead of plasma biomarkers did not improve the accuracy significantly. The clinical predictions by memory clinic physicians had significantly lower accuracy (4-year AUC = 0.71). In summary, plasma P-tau, in combination with brief cognitive tests and APOE genotyping, might greatly improve the diagnostic prediction of AD and facilitate recruitment for AD trials.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Nicholas Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, 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, London, UK.,UK Dementia Research Institute at University College London, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | | | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
68
|
Novak P, Kovacech B, Katina S, Schmidt R, Scheltens P, Kontsekova E, Ropele S, Fialova L, Kramberger M, Paulenka-Ivanovova N, Smisek M, Hanes J, Stevens E, Kovac A, Sutovsky S, Parrak V, Koson P, Prcina M, Galba J, Cente M, Hromadka T, Filipcik P, Piestansky J, Samcova M, Prenn-Gologranc C, Sivak R, Froelich L, Fresser M, Rakusa M, Harrison J, Hort J, Otto M, Tosun D, Ondrus M, Winblad B, Novak M, Zilka N. ADAMANT: a placebo-controlled randomized phase 2 study of AADvac1, an active immunotherapy against pathological tau in Alzheimer's disease. NATURE AGING 2021; 1:521-534. [PMID: 37117834 DOI: 10.1038/s43587-021-00070-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/28/2021] [Indexed: 04/30/2023]
Abstract
Alzheimer's disease (AD) pathology is partly characterized by accumulation of aberrant forms of tau protein. Here we report the results of ADAMANT, a 24-month double-blinded, parallel-arm, randomized phase 2 multicenter placebo-controlled trial of AADvac1, an active peptide vaccine designed to target pathological tau in AD (EudraCT 2015-000630-30). Eleven doses of AADvac1 were administered to patients with mild AD dementia at 40 μg per dose over the course of the trial. The primary objective was to evaluate the safety and tolerability of long-term AADvac1 treatment. The secondary objectives were to evaluate immunogenicity and efficacy of AADvac1 treatment in slowing cognitive and functional decline. A total of 196 patients were randomized 3:2 between AADvac1 and placebo. AADvac1 was safe and well tolerated (AADvac1 n = 117, placebo n = 79; serious adverse events observed in 17.1% of AADvac1-treated individuals and 24.1% of placebo-treated individuals; adverse events observed in 84.6% of AADvac1-treated individuals and 81.0% of placebo-treated individuals). The vaccine induced high levels of IgG antibodies. No significant effects were found in cognitive and functional tests on the whole study sample (Clinical Dementia Rating-Sum of the Boxes scale adjusted mean point difference -0.360 (95% CI -1.306, 0.589)), custom cognitive battery adjusted mean z-score difference of 0.0008 (95% CI -0.169, 0.172). We also present results from exploratory and post hoc analyses looking at relevant biomarkers and clinical outcomes in specific subgroups. Our results show that AADvac1 is safe and immunogenic, but larger stratified studies are needed to better evaluate its potential clinical efficacy and impact on disease biomarkers.
Collapse
Affiliation(s)
- Petr Novak
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia.
| | | | | | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University Graz, Graz, Austria
| | - Philip Scheltens
- Alzheimer Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Stefan Ropele
- Clinical Division of General Neurology, Department of Neurology, Medical University Graz, Graz, Austria
| | | | - Milica Kramberger
- Department of Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | | | - Jozef Hanes
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Eva Stevens
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Andrej Kovac
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Stanislav Sutovsky
- 1st Department of Neurology, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | | | - Peter Koson
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Michal Prcina
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | | | - Martin Cente
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Tomas Hromadka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | - Maria Samcova
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | | | - Roman Sivak
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Lutz Froelich
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit, Medical Faculty Mannheim University of Heidelberg, Heidelberg, Germany
| | | | - Martin Rakusa
- Department of Neurological Diseases, University Medical Centre Maribor, Maribor, Slovenia
| | - John Harrison
- Alzheimer Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Matej Ondrus
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Bengt Winblad
- Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | | | - Norbert Zilka
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| |
Collapse
|
69
|
Dubois B, Villain N, Frisoni GB, Rabinovici GD, Sabbagh M, Cappa S, Bejanin A, Bombois S, Epelbaum S, Teichmann M, Habert MO, Nordberg A, Blennow K, Galasko D, Stern Y, Rowe CC, Salloway S, Schneider LS, Cummings JL, Feldman HH. Clinical diagnosis of Alzheimer's disease: recommendations of the International Working Group. Lancet Neurol 2021; 20:484-496. [PMID: 33933186 PMCID: PMC8339877 DOI: 10.1016/s1474-4422(21)00066-1] [Citation(s) in RCA: 498] [Impact Index Per Article: 124.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/21/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
In 2018, the US National Institute on Aging and the Alzheimer's Association proposed a purely biological definition of Alzheimer's disease that relies on biomarkers. Although the intended use of this framework was for research purposes, it has engendered debate and challenges regarding its use in everyday clinical practice. For instance, cognitively unimpaired individuals can have biomarker evidence of both amyloid β and tau pathology but will often not develop clinical manifestations in their lifetime. Furthermore, a positive Alzheimer's disease pattern of biomarkers can be observed in other brain diseases in which Alzheimer's disease pathology is present as a comorbidity. In this Personal View, the International Working Group presents what we consider to be the current limitations of biomarkers in the diagnosis of Alzheimer's disease and, on the basis of this evidence, we propose recommendations for how biomarkers should and should not be used for diagnosing Alzheimer's disease in a clinical setting. We recommend that Alzheimer's disease diagnosis be restricted to people who have positive biomarkers together with specific Alzheimer's disease phenotypes, whereas biomarker-positive cognitively unimpaired individuals should be considered only at-risk for progression to Alzheimer's disease.
Collapse
Affiliation(s)
- Bruno Dubois
- Assistance Publique-Hôpitaux de Paris (AP-HP) Department of Neurology, Sorbonne University, Paris, France; Institut du Cerveau, Sorbonne University, Paris, France.
| | - Nicolas Villain
- Assistance Publique-Hôpitaux de Paris (AP-HP) Department of Neurology, Sorbonne University, Paris, France; Institut du Cerveau, Sorbonne University, Paris, France
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland; Memory Clinic, University Hospital of Geneva, Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), Saint John of God Clinical Research Centre, Brescia, Italy
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology and Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Marwan Sabbagh
- Cleveland Clinic, Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Stefano Cappa
- University School for Advanced Studies, Pavia, Italy; RCCS Mondino Foundation, Pavia, Italy
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain; Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Stéphanie Bombois
- Assistance Publique-Hôpitaux de Paris (AP-HP) Department of Neurology, Sorbonne University, Paris, France; INSERM, CHU Lille, U1171 - Degenerative and vascular cognitive disorders, University of Lille, Lille, France
| | - Stéphane Epelbaum
- Assistance Publique-Hôpitaux de Paris (AP-HP) Department of Neurology, Sorbonne University, Paris, France; Inria ARAMIS project team, Inria-APHP collaboratio, Sorbonne University, Paris, France; Institut du Cerveau, Sorbonne University, Paris, France
| | - Marc Teichmann
- Assistance Publique-Hôpitaux de Paris (AP-HP) Department of Neurology, Sorbonne University, Paris, France
| | - Marie-Odile Habert
- AP-HP Department of Nuclear Medicine, Sorbonne University, Paris, France; CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Sorbonne University, Paris, France; Institut du Cerveau, Sorbonne University, Paris, France
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institute, Stockholm, Sweden; Theme Aging, The Aging Brain, Karolinska University Hospital, Stockholm, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Douglas Galasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Yaakov Stern
- Cognitive Neuroscience Division, Department of Neurology, Columbia University, New York, NY, USA
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen Salloway
- Department of Neurology and Department of Psychiatry, Alpert Medical School of Brown University, Providence, RI, USA; Butler Hospital, Providence, RI, USA
| | - Lon S Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Jeffrey L Cummings
- Cleveland Clinic, Lou Ruvo Center for Brain Health, Las Vegas, NV, USA; Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Howard H Feldman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Shiley-Marcos Alzheimer's Disease Research Center, University of California San Diego, La Jolla, CA, USA; Alzheimer Disease Cooperative Study, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
70
|
Guo Y, Huang YY, Shen XN, Chen SD, Hu H, Wang ZT, Tan L, Yu JT. Characterization of Alzheimer's tau biomarker discordance using plasma, CSF, and PET. Alzheimers Res Ther 2021; 13:93. [PMID: 33947453 PMCID: PMC8094494 DOI: 10.1186/s13195-021-00834-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/21/2021] [Indexed: 12/03/2022]
Abstract
BACKGROUND We aimed to investigate the tau biomarker discrepancies of Alzheimer's disease (AD) using plasma tau phosphorylated at threonine 181 (p-tau181), cerebrospinal fluid (CSF) p-tau181, and AV1451 positron emission tomography (PET). METHODS In the Alzheimer's Disease Neuroimaging Initiative, 724 non-demented participants were categorized into plasma/CSF and plasma/PET groups. Demographic and clinical variables, amyloid-β (Aβ) burden, flortaucipir-PET binding in Braak regions of interest (ROIs), longitudinal changes in clinical outcomes, and conversion risk were compared. RESULTS Across different tau biomarker groups, the proportion of participants with a discordant profile varied (plasma+/CSF- 15.6%, plasma-/CSF+ 15.3%, plasma+/PET- 22.4%, and plasma-/PET+ 6.1%). Within the plasma/CSF categories, we found an increase from concordant-negative to discordant to concordant-positive in the frequency of Aβ pathology or cognitive impairment, rates of cognitive decline, and risk of cognitive conversion. However, the two discordant categories (plasma+/CSF- and plasma-/CSF+) showed comparable performances, resulting in similarly reduced cognitive capacities. Regarding plasma/PET categories, as expected, PET-positive individuals had increased Aβ burden, elevated flortaucipir retention in Braak ROIs, and accelerated cognitive deterioration than concordant-negative persons. Noteworthy, discordant participants with normal PET exhibited reduced flortaucipir uptake in Braak stage ROIs and slower rates of cognitive decline, relative to those PET-positive. Therefore, individuals with PET abnormality appeared to have advanced tau pathological changes and poorer cognitive function, regardless of the plasma status. Furthermore, these results were found only in individuals with Aβ pathology. CONCLUSIONS Our results indicate that plasma and CSF p-tau181 abnormalities associated with amyloidosis occur simultaneously in the progression of AD pathogenesis and related cognitive decline, before tau-PET turns positive.
Collapse
Affiliation(s)
- Yu Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu-Yuan Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
71
|
Salvadó G, Milà‐Alomà M, Shekari M, Minguillon C, Fauria K, Niñerola‐Baizán A, Perissinotti A, Kollmorgen G, Buckley C, Farrar G, Zetterberg H, Blennow K, Suárez‐Calvet M, Molinuevo JL, Gispert JD. Cerebral amyloid-β load is associated with neurodegeneration and gliosis: Mediation by p-tau and interactions with risk factors early in the Alzheimer's continuum. Alzheimers Dement 2021; 17:788-800. [PMID: 33663013 PMCID: PMC8252618 DOI: 10.1002/alz.12245] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/06/2020] [Accepted: 10/24/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The association between cerebral amyloid-β accumulation and downstream CSF biomarkers is not fully understood, particularly in asymptomatic stages. METHODS In 318 cognitively unimpaired participants, we assessed the association between amyloid-β PET (Centiloid), and cerebrospinal fluid (CSF) biomarkers of several pathophysiological pathways. Interactions by Alzheimer's disease risk factors (age, sex and APOE-ε4), and the mediation effect of tau and neurodegeneration were also investigated. RESULTS Centiloids were positively associated with CSF biomarkers of tau pathology (p-tau), neurodegeneration (t-tau, NfL), synaptic dysfunction (neurogranin) and neuroinflammation (YKL-40, GFAP, sTREM2), presenting interactions with age (p-tau, t-tau, neurogranin) and sex (sTREM2, NfL). Most of these associations were mediated by p-tau, except for NfL. The interaction between sex and amyloid-β on sTREM2 and NfL was also tau-independent. DISCUSSION Early amyloid-β accumulation has a tau-independent effect on neurodegeneration and a tau-dependent effect on neuroinflammation. Besides, sex has a modifier effect on these associations independent of tau.
Collapse
Affiliation(s)
- Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | - Marta Milà‐Alomà
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Aida Niñerola‐Baizán
- Nuclear Medicine DepartmentHospital Clínic BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, (CIBER‐BBN)BarcelonaSpain
| | - Andrés Perissinotti
- Nuclear Medicine DepartmentHospital Clínic BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, (CIBER‐BBN)BarcelonaSpain
| | | | | | | | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, (CIBER‐BBN)BarcelonaSpain
| | | |
Collapse
|
72
|
Schwarz AJ. The Use, Standardization, and Interpretation of Brain Imaging Data in Clinical Trials of Neurodegenerative Disorders. Neurotherapeutics 2021; 18:686-708. [PMID: 33846962 PMCID: PMC8423963 DOI: 10.1007/s13311-021-01027-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Imaging biomarkers play a wide-ranging role in clinical trials for neurological disorders. This includes selecting the appropriate trial participants, establishing target engagement and mechanism-related pharmacodynamic effect, monitoring safety, and providing evidence of disease modification. In the early stages of clinical drug development, evidence of target engagement and/or downstream pharmacodynamic effect-especially with a clear relationship to dose-can provide confidence that the therapeutic candidate should be advanced to larger and more expensive trials, and can inform the selection of the dose(s) to be further tested, i.e., to "de-risk" the drug development program. In these later-phase trials, evidence that the therapeutic candidate is altering disease-related biomarkers can provide important evidence that the clinical benefit of the compound (if observed) is grounded in meaningful biological changes. The interpretation of disease-related imaging markers, and comparability across different trials and imaging tools, is greatly improved when standardized outcome measures are defined. This standardization should not impinge on scientific advances in the imaging tools per se but provides a common language in which the results generated by these tools are expressed. PET markers of pathological protein aggregates and structural imaging of brain atrophy are common disease-related elements across many neurological disorders. However, PET tracers for pathologies beyond amyloid β and tau are needed, and the interpretability of structural imaging can be enhanced by some simple considerations to guard against the possible confound of pseudo-atrophy. Learnings from much-studied conditions such as Alzheimer's disease and multiple sclerosis will be beneficial as the field embraces rarer diseases.
Collapse
Affiliation(s)
- Adam J Schwarz
- Takeda Pharmaceuticals Ltd., 40 Landsdowne Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
73
|
Buckley RF. Recent Advances in Imaging of Preclinical, Sporadic, and Autosomal Dominant Alzheimer's Disease. Neurotherapeutics 2021; 18:709-727. [PMID: 33782864 PMCID: PMC8423933 DOI: 10.1007/s13311-021-01026-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Observing Alzheimer's disease (AD) pathological changes in vivo with neuroimaging provides invaluable opportunities to understand and predict the course of disease. Neuroimaging AD biomarkers also allow for real-time tracking of disease-modifying treatment in clinical trials. With recent neuroimaging advances, along with the burgeoning availability of longitudinal neuroimaging data and big-data harmonization approaches, a more comprehensive evaluation of the disease has shed light on the topographical staging and temporal sequencing of the disease. Multimodal imaging approaches have also promoted the development of data-driven models of AD-associated pathological propagation of tau proteinopathies. Studies of autosomal dominant, early sporadic, and late sporadic courses of the disease have shed unique insights into the AD pathological cascade, particularly with regard to genetic vulnerabilities and the identification of potential drug targets. Further, neuroimaging markers of b-amyloid, tau, and neurodegeneration have provided a powerful tool for validation of novel fluid cerebrospinal and plasma markers. This review highlights some of the latest advances in the field of human neuroimaging in AD across these topics, particularly with respect to positron emission tomography and structural and functional magnetic resonance imaging.
Collapse
Affiliation(s)
- Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital & Brigham and Women's, Harvard Medical School, Boston, MA, USA.
- Melbourne School of Psychological Sciences and Florey Institutes, University of Melbourne, Melbourne, VIC, Australia.
- Department of Neurology, Massachusetts General Hospital, 149 13th St, Charlestown, MA, 02129, USA.
| |
Collapse
|
74
|
Leuzy A, Cullen NC, Mattsson-Carlgren N, Hansson O. Current advances in plasma and cerebrospinal fluid biomarkers in Alzheimer's disease. Curr Opin Neurol 2021; 34:266-274. [PMID: 33470669 DOI: 10.1097/wco.0000000000000904] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review provides a concise overview of recent advances in cerebrospinal fluid (CSF) and blood-based biomarkers of Alzheimer's disease lesions. RECENT FINDINGS Important recent advances for CSF Alzheimer's disease biomarkers include the introduction of fully automated assays, the development and implementation of certified reference materials for CSF Aβ42 and a unified protocol for handling of samples, which all support reliability and availability of CSF Alzheimer's disease biomarkers. Aβ deposition can be detected using Aβ42/Aβ40 ratio in both CSF and plasma, though a much more modest change is seen in plasma. Tau aggregation can be detected using phosphorylated tau (P-tau) at threonine 181 and 217 in CSF, with similar accuracy in plasma. Neurofilament light (NfL) be measured in CSF and shows similar diagnostic accuracy in plasma. Though total tau (T-tau) can also be measured in plasma, this measure is of limited clinical relevance for Alzheimer's disease in its current immunoassay format. SUMMARY Alzheimer's disease biomarkers, including Aβ, P-tau and NfL can now be reliably measured in both CSF and blood. Plasma-based measures of P-tau show particular promise, with potential applications in both clinical practice and in clinical trials.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
- Department of Neurology, Skåne University Hospital
- Wallenberg Centre for Molecular Medicine, Lund University
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
75
|
O'Shea DM, Thomas KR, Asken B, Lee AK, Davis JD, Malloy PF, Salloway SP, Correia S. Adding cognition to AT(N) models improves prediction of cognitive and functional decline. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12174. [PMID: 33816757 PMCID: PMC8012408 DOI: 10.1002/dad2.12174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION This study sought to determine whether adding cognition to a model with Alzheimer's disease biomarkers based on the amyloid, tau, and neurodegeneration/neuronal injury-AT(N)-biomarker framework predicts rates of cognitive and functional decline in older adults without dementia. METHODS The study included 465 participants who completed amyloid positron emission tomography, cerebrospinal fluid phosphorylated tau, structural magnetic resonance imaging, and serial neuropsychological testing. Using the AT(N) framework and a newly validated cognitive metric as the independent variables, we used linear mixed effects models to examine a 4-year rate of change in cognitive and functional measures. RESULTS The inclusion of baseline cognitive status improved model fit in predicting rate of decline in outcomes above and beyond biomarker variables. Specifically, those with worse cognitive functioning at baseline had faster rates of memory and functional decline over a 4-year period, even when accounting for AT(N). DISCUSSION Including a newly validated measure of baseline cognition may improve clinical prognosis in non-demented older adults beyond the use of AT(N) biomarkers alone.
Collapse
Affiliation(s)
- Deirdre M. O'Shea
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Kelsey R. Thomas
- Research Service, VA San Diego Healthcare SystemUniversity of California San DiegoSan DiegoCaliforniaUSA
- Department of PsychiatryUniversity of California, San Diego, La JollaCAUSA
| | - Breton Asken
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Athene K.W. Lee
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Jennifer D. Davis
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Paul F. Malloy
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Stephen P. Salloway
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Stephen Correia
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | | |
Collapse
|
76
|
Shi L, Winchester LM, Westwood S, Baird AL, Anand SN, Buckley NJ, Hye A, Ashton NJ, Bos I, Vos SJB, Kate MT, Scheltens P, Teunissen CE, Vandenberghe R, Gabel S, Meersmans K, Engelborghs S, De Roeck EE, Sleegers K, Frisoni GB, Blin O, Richardson JC, Bordet R, Molinuevo JL, Rami L, Wallin A, Kettunen P, Tsolaki M, Verhey F, Lléo A, Sala I, Popp J, Peyratout G, Martinez-Lage P, Tainta M, Johannsen P, Freund-Levi Y, Frölich L, Dobricic V, Legido-Quigley C, Barkhof F, Andreasson U, Blennow K, Zetterberg H, Streffer J, Lill CM, Bertram L, Visser PJ, Kolb HC, Narayan VA, Lovestone S, Nevado-Holgado AJ. Replication study of plasma proteins relating to Alzheimer's pathology. Alzheimers Dement 2021; 17:1452-1464. [PMID: 33792144 DOI: 10.1002/alz.12322] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/26/2020] [Accepted: 02/05/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION This study sought to discover and replicate plasma proteomic biomarkers relating to Alzheimer's disease (AD) including both the "ATN" (amyloid/tau/neurodegeneration) diagnostic framework and clinical diagnosis. METHODS Plasma proteins from 972 subjects (372 controls, 409 mild cognitive impairment [MCI], and 191 AD) were measured using both SOMAscan and targeted assays, including 4001 and 25 proteins, respectively. RESULTS Protein co-expression network analysis of SOMAscan data revealed the relation between proteins and "N" varied across different neurodegeneration markers, indicating that the ATN variants are not interchangeable. Using hub proteins, age, and apolipoprotein E ε4 genotype discriminated AD from controls with an area under the curve (AUC) of 0.81 and MCI convertors from non-convertors with an AUC of 0.74. Targeted assays replicated the relation of four proteins with the ATN framework and clinical diagnosis. DISCUSSION Our study suggests that blood proteins can predict the presence of AD pathology as measured in the ATN framework as well as clinical diagnosis.
Collapse
Affiliation(s)
- Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | | | - Sarah Westwood
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Alison L Baird
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Sneha N Anand
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Abdul Hye
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Nicholas J Ashton
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK.,Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Isabelle Bos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands.,Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Mara Ten Kate
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Philip Scheltens
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry lab, Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | | | - Silvy Gabel
- University Hospital Leuven, Leuven, Belgium.,Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Karen Meersmans
- University Hospital Leuven, Leuven, Belgium.,Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology, UZ Brussel and Center for Neurociences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ellen E De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Kristel Sleegers
- Complex Genetics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.,Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Giovanni B Frisoni
- University of Geneva, Geneva, Switzerland.,IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Olivier Blin
- AIX marseille university, INS, Ap-hm, Marseille, France
| | | | - Régis Bordet
- Inserm, University of Lille, CHU Lille, Lille, France
| | - José L Molinuevo
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hopsital Clínic-IDIBAPS, Barcelona, Spain.,Barcelona Beta Brain Research Center, Unversitat Pompeu Fabra, Barcelona, Spain
| | - Lorena Rami
- Barcelona Beta Brain Research Center, Unversitat Pompeu Fabra, Barcelona, Spain
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, Makedonia, Thessaloniki, Greece
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Alberto Lléo
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Isabel Sala
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julius Popp
- University Hospital of Lausanne, Lausanne, Switzerland.,Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | | | | | | | - Peter Johannsen
- Danish Dementia Research Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Yvonne Freund-Levi
- Maurice Wohl Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK.,Karolinska Institutet Center for Alzheimer Research, Division of Clinical Geriatrics, School of Medical Sciences Örebro University and Department of Neurobiology, Caring Sciences and Society (NVS), Stockholm, Sweden
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit, University of Heidelberg, Mannheim, Germany
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Cristina Legido-Quigley
- Kings College London, London, UK.,The Systems Medicine Group, Steno Diabetes Center, Gentofte, Denmark
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherland.,UCL Institutes of Neurology and Healthcare Engineering, London, UK
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Johannes Streffer
- Complex Genetics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.,UCB, Braine-l'Alleud, Belgium, formerly Janssen R&D, LLC Beerse, Beerse, Belgium
| | - Christina M Lill
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.,Ageing Epidemiology Research Unit, School of Public Health, Imperial College, London, UK
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany.,Department of Psychology, University of Oslo, Oslo, Norway
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands.,Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, UK.,Janssen R&D, Beerse, UK
| | | |
Collapse
|
77
|
Altiné‐Samey R, Antier D, Mavel S, Dufour‐Rainfray D, Balageas A, Beaufils E, Emond P, Foucault‐Fruchard L, Chalon S. The contributions of metabolomics in the discovery of new therapeutic targets in Alzheimer's disease. Fundam Clin Pharmacol 2021; 35:582-594. [DOI: 10.1111/fcp.12654] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/05/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Affiliation(s)
| | - Daniel Antier
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service Pharmacie Tours France
| | - Sylvie Mavel
- UMR 1253 iBrain Université de Tours Inserm, Tours France
| | - Diane Dufour‐Rainfray
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service de Médecine Nucléaire In Vitro Tours France
| | | | - Emilie Beaufils
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Centre Mémoire Ressources et Recherche Tours France
| | - Patrick Emond
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service de Médecine Nucléaire In Vitro Tours France
| | - Laura Foucault‐Fruchard
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service Pharmacie Tours France
| | - Sylvie Chalon
- UMR 1253 iBrain Université de Tours Inserm, Tours France
| |
Collapse
|
78
|
Therriault J, Pascoal TA, Benedet AL, Tissot C, Savard M, Chamoun M, Lussier F, Kang MS, Berzgin G, Wang T, Fernandes-Arias J, Massarweh G, Soucy JP, Vitali P, Saha-Chaudhuri P, Gauthier S, Rosa-Neto P. Frequency of Biologically Defined Alzheimer Disease in Relation to Age, Sex, APOE ε4, and Cognitive Impairment. Neurology 2021; 96:e975-e985. [PMID: 33443136 PMCID: PMC8055338 DOI: 10.1212/wnl.0000000000011416] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To assess the frequency of biologically defined Alzheimer disease (AD) in relation to age, sex, APOE ε4, and clinical diagnosis in a prospective cohort study evaluated with amyloid-PET and tau-PET. METHODS We assessed cognitively unimpaired (CU) elderly (n = 166), patients with amnestic mild cognitive impairment (n = 77), and patients with probable AD dementia (n = 62) who underwent evaluation by dementia specialists and neuropsychologists in addition to amyloid-PET with [18F]AZD4694 and tau-PET with [18F]MK6240. Individuals were grouped according to their AD biomarker profile. Positive predictive value for biologically defined AD was assessed in relation to clinical diagnosis. Frequency of AD biomarker profiles was assessed using logistic regressions with odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The clinical diagnosis of probable AD dementia demonstrated good agreement with biologically defined AD (positive predictive value 85.2%). A total of 7.88% of CU were positive for both amyloid-PET and tau-PET. Frequency of biologically defined AD increased with age (OR 1.14; p < 0.0001) and frequency of APOE ε4 allele carriers (single ε4: OR 3.82; p < 0.0001; double ε4: OR 17.55, p < 0.0001). CONCLUSION Whereas we observed strong, but not complete, agreement between clinically defined probable AD dementia and biomarker positivity for both β-amyloid and tau, we also observed that biologically defined AD was not rare in CU elderly. Abnormal tau-PET was almost exclusively observed in individuals with abnormal amyloid-PET. Our results highlight that even in tertiary care memory clinics, detailed evaluation by dementia specialists systematically underestimates the frequency of biologically defined AD and related entities. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that biologically defined AD (abnormal amyloid PET and tau PET) was observed in 85.2% of people with clinically defined AD and 7.88% of CU elderly.
Collapse
Affiliation(s)
- Joseph Therriault
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Tharick A Pascoal
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Andrea L Benedet
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Cecile Tissot
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Melissa Savard
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Mira Chamoun
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Firoza Lussier
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Min Su Kang
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Gleb Berzgin
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Tina Wang
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Jaime Fernandes-Arias
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Gassan Massarweh
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Jean-Paul Soucy
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Paolo Vitali
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Paramita Saha-Chaudhuri
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Serge Gauthier
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada
| | - Pedro Rosa-Neto
- From the Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital (J.T., T.A.P., A.L.B., C.T., M.S., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., S.G., P.R.-N.), and the Departments of Neurology and Neurosurgery (J.T., T.A.P., A.L.B., C.T., M.C., F.L., M.S.K., G.B., T.W., J.F.-A., J.-P.S., P.V., S.G., P.R.-N.), Psychiatry (S.G., P.R.-N.), Radiochemistry (G.M.), and Epidemiology and Biostatistics (P.S.-C.), McGill University, Montreal; and Montreal Neurological Institute (J.T., T.A.P., A.L.B., C.T., F.L., M.S.K., G.B., T.W., J.F.-A., G.M., J.-P.S., P.R.-N.), Canada.
| |
Collapse
|
79
|
Provost K, Iaccarino L, Soleimani-Meigooni DN, Baker S, Edwards L, Eichenlaub U, Hansson O, Jagust W, Janabi M, La Joie R, Lesman-Segev O, Mellinger TJ, Miller BL, Ossenkoppele R, Pham J, Smith R, Sonni I, Strom A, Mattsson-Carlgren N, Rabinovici GD. Comparing ATN-T designation by tau PET visual reads, tau PET quantification, and CSF PTau181 across three cohorts. Eur J Nucl Med Mol Imaging 2021; 48:2259-2271. [PMID: 33398408 DOI: 10.1007/s00259-020-05152-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/06/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE To compare rates of tau biomarker positivity (T-status) per the 2018 Alzheimer's Disease (AD) Research Framework derived from [18F]flortaucipir (FTP) PET visual assessment, FTP quantification, and cerebrospinal fluid (CSF) phosphorylated Tau-181 (PTau181). METHODS We included 351 subjects with varying clinical diagnoses from three cohorts with available FTP PET and CSF PTau181 within 18 months. T-status was derived from (1) FTP visual assessment by two blinded raters; (2) FTP standardized uptake value ratio (SUVR) quantification from a temporal meta-ROI (threshold: SUVR ≥1.27); and (3) Elecsys® Phospho-Tau (181P) CSF (Roche Diagnostics) concentrations (threshold: PTau181 ≥ 24.5 pg/mL). RESULTS FTP visual reads yielded the highest rates of T+, while T+ by SUVR increased progressively from cognitively normal (CN) through mild cognitive impairment (MCI) and AD dementia. T+ designation by CSF PTau181 was intermediate between FTP visual reads and SUVR values in CN, similar to SUVR in MCI, and lower in AD dementia. Concordance in T-status between modality pairs ranged from 68 to 76% and varied by clinical diagnosis, being highest in patients with AD dementia. In discriminating Aβ + MCI and AD subjects from healthy controls and non-AD participants, FTP visual assessment was most sensitive (0.96) but least specific (0.60). Specificity was highest with FTP SUVR (0.91) with sensitivity of 0.89. Sensitivity (0.73) and specificity (0.72) were balanced for PTau181. CONCLUSION The choice of tau biomarker may differ by disease stage and research goals that seek to maximize sensitivity or specificity. Visual interpretations of tau PET enhance sensitivity compared to quantification alone, particularly in early disease stages.
Collapse
Affiliation(s)
- Karine Provost
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA.
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Suzanne Baker
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | | | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - William Jagust
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA, USA
| | - Mustafa Janabi
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Orit Lesman-Segev
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Taylor J Mellinger
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Julie Pham
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Ida Sonni
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, UC Los Angeles, Los Angeles, CA, USA
| | - Amelia Strom
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94143, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
80
|
Alzheimer's disease profiled by fluid and imaging markers: tau PET best predicts cognitive decline. Mol Psychiatry 2021; 26:5888-5898. [PMID: 34593971 PMCID: PMC8758489 DOI: 10.1038/s41380-021-01263-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/23/2021] [Accepted: 08/04/2021] [Indexed: 11/08/2022]
Abstract
For early detection of Alzheimer's disease, it is important to find biomarkers with predictive value for disease progression and clinical manifestations, such as cognitive decline. Individuals can now be profiled based on their biomarker status for Aβ42 (A) or tau (T) deposition and neurodegeneration (N). The aim of this study was to compare the cerebrospinal fluid (CSF) and imaging (PET/MR) biomarkers in each ATN category and to assess their ability to predict longitudinal cognitive decline. A subset of 282 patients, who had had at the same time PET investigations with amyloid-β and tau tracers, CSF sampling, and structural MRI (18% within 13 months), was selected from the ADNI dataset. The participants were grouped by clinical diagnosis at that time: cognitively normal, subjective memory concern, early or late mild cognitive impairment, or AD. Agreement between CSF (amyloid-β-1-42(A), phosphorylated-Tau181(T), total-Tau(N)), and imaging (amyloid-β PET (florbetaben and florbetapir)(A), tau PET (flortaucipir)(T), hippocampal volume (MRI)(N)) positivity in ATN was assessed with Cohen's Kappa. Linear mixed-effects models were used to predict decline in the episodic memory. There was moderate agreement between PET and CSF for A biomarkers (Kappa = 0.39-0.71), while only fair agreement for T biomarkers (Kappa ≤ 0.40, except AD) and discordance for N biomarkers across all groups (Kappa ≤ 0.14) was found. Baseline PET tau predicted longitudinal decline in episodic memory irrespective of CSF p-Tau181 positivity (p ≤ 0.02). Baseline PET tau and amyloid-β predicted decline in episodic memory (p ≤ 0.0001), but isolated PET amyloid-β did not. Isolated PET Tau positivity was only observed in 2 participants (0.71% of the sample). While results for amyloid-β were similar using CSF or imaging, CSF and imaging results for tau and neurodegeneration were not interchangeable. PET tau positivity was superior to CSF p-Tau181 and PET amyloid-β in predicting cognitive decline in the AD continuum within 3 years of follow-up.
Collapse
|
81
|
Cousins KAQ, Phillips JS, Irwin DJ, Lee EB, Wolk DA, Shaw LM, Zetterberg H, Blennow K, Burke SE, Kinney NG, Gibbons GS, McMillan CT, Trojanowski JQ, Grossman M. ATN incorporating cerebrospinal fluid neurofilament light chain detects frontotemporal lobar degeneration. Alzheimers Dement 2020; 17:822-830. [PMID: 33226735 DOI: 10.1002/alz.12233] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The ATN framework provides an in vivo diagnosis of Alzheimer's disease (AD) using cerebrospinal fluid (CSF) biomarkers of pathologic amyloid plaques (A), tangles (T), and neurodegeneration (N). ATN is rarely evaluated in pathologically confirmed patients and its poor sensitivity to suspected non-Alzheimer's pathophysiologies (SNAP), including frontotemporal lobar degeneration (FTLD), leads to misdiagnoses. We compared accuracy of ATN (ATNTAU ) using CSF total tau (t-tau) to a modified strategy (ATNNfL ) using CSF neurofilament light chain (NfL) in an autopsy cohort. METHODS ATNTAU and ATNNfL were trained in an independent sample and validated in autopsy-confirmed AD (n = 67) and FTLD (n = 27). RESULTS ATNNfL more accurately identified FTLD as SNAP (sensitivity = 0.93, specificity = 0.94) than ATNTAU (sensitivity = 0.44, specificity = 0.97), even in cases with co-occurring AD and FTLD. ATNNfL misclassified fewer AD and FTLD as "Normal" (2%) than ATNTAU (14%). DISCUSSION ATNNfL is a promising diagnostic strategy that may accurately identify both AD and FTLD, even when pathologies co-occur.
Collapse
Affiliation(s)
- Katheryn A Q Cousins
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeffrey S Phillips
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Wolk
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute, University College London, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Sarah E Burke
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nikolas G Kinney
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Garrett S Gibbons
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Corey T McMillan
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
82
|
Guo Y, Li H, Tan L, Chen S, Yang Y, Ma Y, Zuo C, Dong Q, Tan L, Yu J. Discordant Alzheimer's neurodegenerative biomarkers and their clinical outcomes. Ann Clin Transl Neurol 2020; 7:1996-2009. [PMID: 32949193 PMCID: PMC7545611 DOI: 10.1002/acn3.51196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/08/2020] [Accepted: 08/25/2020] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE In the 2018 ATN framework, Alzheimer's neurodegenerative biomarkers comprised cerebrospinal fluid (CSF) total tau, 18 F-fluorodeoxyglucose-positron emission tomography, and brain atrophy. We aimed to assess the clinical outcomes of having discordant Alzheimer's neurodegenerative biomarkers. METHODS A total of 721 non-demented individuals from the Alzheimer's Disease Neuroimaging Initiative database were included and then further categorized into concordant-negative, discordant, and concordant-positive groups. Demographic distributions of the groups were compared. Longitudinal changes in clinical outcomes and risk of conversion were assessed using linear mixed-effects models and multivariate Cox proportional hazard models, respectively. RESULTS Discordant group was intermediate to concordant-negative and concordant-positive groups in terms of APOE ε4 positivity, CSF amyloid-beta, and phosphorylated tau. Compared with concordant-negative group, discordant group deteriorated faster in cognitive scores (Mini-Mental State Examination, the Clinical Dementia Rating Scale-Sum of Boxes, and the Functional Activities Questionnaire) and demonstrated greater rates of atrophy in brain structures (hippocampus, entorhinal cortex, and whole brain), and concordant-positive group performed worse over time than discordant group. Moreover, the risk of cognitive decline increased from concordant-negative to discordant to concordant-positive. The results from longitudinal analyses were validated in A+T+, cognitively normal, and mild cognitive impairment individuals, and were also validated by applying different cutoffs and neurodegenerative biomarkers. INTERPRETATION Discordant neurodegenerative status denotes a stage of cognitive function which is intermediate between concordant-negative and concordant-positive. Identification of discordant cases would provide insights into intervention and new therapy approaches, particularly in A+T+ individuals. Moreover, this work may be a complement to the ATN scheme.
Collapse
Affiliation(s)
- Yu Guo
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Hong‐Qi Li
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Lin Tan
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Shi‐Dong Chen
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Yu‐Xiang Yang
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Ya‐Hui Ma
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Chuan‐Tao Zuo
- PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Qiang Dong
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Lan Tan
- Department of NeurologyQingdao Municipal Hospital Affiliated to Qingdao UniversityQingdaoChina
| | - Jin‐Tai Yu
- Department of Neurology and Institute of NeurologyHuashan Hospital, Shanghai Medical College, Fudan UniversityShanghaiChina
| | | |
Collapse
|