1
|
Schöll M, Verberk IMW, Del Campo M, Delaby C, Therriault J, Chong JR, Palmqvist S, Alcolea D. Challenges in the practical implementation of blood biomarkers for Alzheimer's disease. THE LANCET. HEALTHY LONGEVITY 2024; 5:100630. [PMID: 39369727 DOI: 10.1016/j.lanhl.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/23/2024] [Accepted: 07/29/2024] [Indexed: 10/08/2024] Open
Abstract
Blood biomarkers have emerged as accessible, cost-effective, and highly promising tools for advancing the diagnostics of Alzheimer's disease. However, transitioning from cerebrospinal fluid biomarkers to blood biomarkers-eg, to verify amyloid β pathology-requires careful consideration. This Series paper highlights the main challenges in the implementation of blood biomarkers for Alzheimer's disease in different possible contexts of use. Despite the robustness of measuring blood biomarker concentrations, the widespread adoption of blood biomarkers requires rigorous standardisation efforts to address inherent challenges in diverse contexts of use. The challenges include understanding the effect of pre-analytical and analytical conditions, potential confounding factors, and comorbidities that could influence outcomes of blood biomarkers and their use in diverse populations. Additionally, distinct scenarios present their own specific challenges. In memory clinics, the successful integration of blood biomarkers in diagnostic tests will require well-established diagnostic accuracy and comprehensive assessments of the effect of blood biomarkers on the diagnostic confidence and patient management of clinicians. In primary care settings, and even more when implemented in population-based screening programmes for which no experience with any biomarkers for Alzheimer's disease currently exists, the implementation of blood biomarkers will be challenged by the need for education of primary care clinical staff and clear guidelines. However, despite the challenges, blood biomarkers hold great promise for substantially enhancing the diagnostic accuracy and effectively streamlining referral processes, leading to earlier diagnosis and access to treatments. The ongoing efforts that are shaping the integration of blood biomarkers across diverse clinical settings pave the way towards precision medicine in Alzheimer's disease.
Collapse
Affiliation(s)
- Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden; Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Marta Del Campo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute (IMIM), Barcelona, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Constance Delaby
- LBPC-PPC, University of Montpellier, CHU Montpellier, INM INSERM, Montpellier, France; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada; Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health Systems, Singapore
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Clinical Sciences in Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
2
|
Wuestefeld A, Pichet Binette A, van Westen D, Strandberg O, Stomrud E, Mattsson-Carlgren N, Janelidze S, Smith R, Palmqvist S, Baumeister H, Berron D, Yushkevich PA, Hansson O, Spotorno N, Wisse LEM. Medial temporal lobe atrophy patterns in early-versus late-onset amnestic Alzheimer's disease. Alzheimers Res Ther 2024; 16:204. [PMID: 39285454 PMCID: PMC11403779 DOI: 10.1186/s13195-024-01571-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND The medial temporal lobe (MTL) is hypothesized to be relatively spared in early-onset Alzheimer's disease (EOAD). Yet, detailed examination of MTL subfields and drivers of atrophy in amnestic EOAD is lacking. METHODS BioFINDER-2 participants with memory impairment, abnormal amyloid-β and tau-PET were included. Forty-one amnestic EOAD individuals ≤65 years and, as comparison, late-onset AD (aLOAD, ≥70 years, n = 154) and amyloid-β-negative cognitively unimpaired controls were included. MTL subregions and biomarkers of (co-)pathologies were measured. RESULTS AD groups showed smaller MTL subregions compared to controls. Atrophy patterns were similar across AD groups: aLOAD showed thinner entorhinal cortices than aEOAD; aEOAD showed thinner parietal regions than aLOAD. aEOAD showed lower white matter hyperintensities than aLOAD. No differences in MTL tau-PET or transactive response DNA binding protein 43-proxy positivity were found. CONCLUSIONS We found evidence for MTL atrophy in amnestic EOAD and overall similar levels to aLOAD of MTL tau pathology and co-pathologies.
Collapse
Affiliation(s)
- Anika Wuestefeld
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden.
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
| | - Danielle van Westen
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, Klinikgatan 13B, Lund, SE-22242, Sweden
- Image and Function, Skåne University Hospital, Lund, 22242, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 20502, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
- Department of Neurology, Skåne University Hospital, Lund, 22242, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, 22184, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 20502, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 20502, Sweden
| | - Hannah Baumeister
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany
| | - Paul A Yushkevich
- Penn Image Computing and Science Laboratory (PICSL), Department of Radiology, University of Pennsylvania, Philadelphia, 19104, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, 20502, Sweden
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Klinikgatan 28, Room C1103b, Lund, SE-22242, Sweden
| | - Laura E M Wisse
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, Klinikgatan 13B, Lund, SE-22242, Sweden.
| |
Collapse
|
3
|
Pichet Binette A, Mammana A, Wisse L, Rossi M, Strandberg O, Smith R, Mattsson-Carlgren N, Janelidze S, Palmqvist S, Ticca A, Stomrud E, Parchi P, Hansson O. Associations between misfolded alpha-synuclein aggregates and Alzheimer's disease pathology in vivo. Alzheimers Dement 2024. [PMID: 39258841 DOI: 10.1002/alz.14225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION We examined the relations of misfolded alpha synuclein (α-synuclein) with Alzheimer's disease (AD) biomarkers in two large independent cohorts. METHODS We included Biomarkers for Identifying Neurodegenerative Disorders Early and Reliably Two (BioFINDER-2) and Alzheimer's Disease Neuroimaging Initiative (ADNI) participants (n = 2315, cognitively unimpaired, mild cognitive impairment, AD dementia) who had cross-sectional cerebrospinal fluid (CSF) α-synuclein measurement from seed-amplification assay as well as cross-sectional and longitudinal amyloid beta (Aβ) and tau levels (measured in CSF and/or by positron emission tomography). All analyses were adjusted for age, sex, and cognitive status. RESULTS Across cohorts, the main biomarker associated with α-synuclein positivity at baseline was higher levels of Aβ pathology (all p values ≤ 0.02), but not tau. Looking at longitudinal measures of AD biomarkers, α-synuclein -positive participants had a statistically significant faster increase of Aβ load, although of modest magnitude (1.11 Centiloid/year, p = 0.02), compared to α-synuclein -negative participants in BioFINDER-2 but not in ADNI. DISCUSSION We showed associations between concurrent misfolded α-synuclein and Aβ levels, providing in vivo evidence of links between these two molecular disease pathways in humans. HIGHLIGHTS Amyloid beta (Aβ), but not tau, was associated with alpha-synuclein (α-synuclein) positivity. Such association was consistent across two cohorts, beyond the effect of age, sex, and cognitive status. α-synuclein-positive participants had a small, statistically significant faster increase in Aβ positron emission tomography levels in one of the two cohorts.
Collapse
Grants
- NIA NIH HHS
- Alzheimer's Association
- 2022-00775 GHR Foundation, Swedish Research Council
- 2021-02219 GHR Foundation, Swedish Research Council
- 2018-02052 GHR Foundation, Swedish Research Council
- ERAPERMED2021-184 ERA PerMed
- 2022-0231 Knut and Alice Wallenberg foundation
- AF-980907 Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson's disease) at Lund University, Swedish Alzheimer Foundation
- AF-980832 Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson's disease) at Lund University, Swedish Alzheimer Foundation
- AF-993465 Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson's disease) at Lund University, Swedish Alzheimer Foundation
- AF-939981 Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson's disease) at Lund University, Swedish Alzheimer Foundation
- Swedish Brain Foundation
- 1412/22 Parkinson foundation of Sweden
- WASP/DDLS22-066 Cure Alzheimer's fund
- 2019-03401 EU Joint Programme Neurodegenerative Diseases
- FRS-0003 Rönström Family Foundation
- Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse
- 2020-O000028 Skåne University Hospital Foundation
- 2022-1259 Regionalt Forskningsstöd
- Swedish federal government under the ALF agreement
- Italian Ministero della Salute
- Alzheimer's Disease Neuroimaging Initiative
- NIH HHS
- NIBIB NIH HHS
- Alzheimer's Drug Discovery Foundation
- Araclon Biotech
- BioClinica, Inc.
- Biogen; BristolMyers Squibb Company
- CereSpir, Inc.
- Cogstate
- Eisai Inc.
- Elan Pharmaceuticals, Inc.
- Eli Lilly and Company; EuroImmun
- F. Hoffmann-LaRoche Ltdand its affiliated company Genentech, Inc.
- Fujirebio
- GE Healthcare
- IXICO Ltd.
- Janssen Alzheimer Immunotherapy Research & Development
- Johnson & Johnson Pharmaceutical Research & Development LLC
- Lumosity; Lundbeck
- Merck & Co., Inc.
- MesoScale Diagnostics
- LLC
- NeuroRxResearch
- Neurotrack Technologies
- Novartis Pharmaceuticals Corporation
- Pfizer Inc.
- Piramal Imaging
- Takeda Pharmaceutical Company; and Transition Therapeutics
- 298314 Fonds de Recherche en Santé Québec
- ADG-101096455 H2020 European Research Council
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Angela Mammana
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Laura Wisse
- Diagnostic Radiology Unit, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Marcello Rossi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Alice Ticca
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
4
|
Römpler K, Arendt P, Brix B, Borchardt-Lohölter V, Schulz A, Busse M, Busse S. Evaluation of the EUROIMMUN automated chemiluminescence immunoassays for measurement of four core biomarkers for Alzheimer's disease in cerebrospinal fluid. Pract Lab Med 2024; 41:e00425. [PMID: 39314784 PMCID: PMC11417521 DOI: 10.1016/j.plabm.2024.e00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Robust immunoassays for quantification of Alzheimer's disease (AD)-specific biomarkers are required for routine diagnostics. We report analytical performance characteristics of four new chemiluminescence immunoassays (ChLIA, EUROIMMUN) running on closed, fully automated random-access instruments for quantification of Aβ1-40, Aβ1-42, tTau, and pTau(181) in human cerebrospinal fluid (CSF). Methods ChLIAs were validated according to the guidelines of the Clinical and Laboratory Standards Institute (CLSI). Optimal cut-offs for biomarkers and biomarker ratios were determined using samples from 219 AD patients and 220 patients with AD-related symptoms. For performance comparison, biomarker concentrations were measured in 110 diagnostic leftover samples using the ChLIAs and established Lumipulse G assays (Fujirebio). Results All ChLIAs met CLSI criteria. Overall agreement between assays was 89.0%-97.3 % with highly correlating results (Pearson's correlation coefficients: 0.82-0.99). Passing-Bablok regression analysis revealed systematic differences. Discussion EUROIMMUN ChLIAs showed good analytical performances and represent new valuable tools for diagnostics of AD.
Collapse
Affiliation(s)
- Katharina Römpler
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Luebeck, Germany
| | - Philipp Arendt
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Luebeck, Germany
| | - Britta Brix
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Luebeck, Germany
| | - Viola Borchardt-Lohölter
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Luebeck, Germany
| | - Anette Schulz
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Luebeck, Germany
| | - Mandy Busse
- Department for Experimental Obstetrics and Gynecology, Otto von Guericke University Magdeburg, Medical Faculty, Gerhart-Hauptmann-Str. 35, 39180, Magdeburg, Germany
- University Hospital for Psychiatry and Psychotherapy, Otto von Guericke University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Stefan Busse
- University Hospital for Psychiatry and Psychotherapy, Otto von Guericke University Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| |
Collapse
|
5
|
Agnello L, Giglio RV, Del Ben F, Piccoli T, Colletti T, Scazzone C, Lo Sasso B, Ciaccio AM, Gambino CM, Salemi G, Ciaccio M. Evaluation of core Biomarkers of Alzheimer's disease in saliva and plasma measured by chemiluminescent enzyme immunoassays on a fully automated platform. Sci Rep 2024; 14:16084. [PMID: 38992063 PMCID: PMC11239824 DOI: 10.1038/s41598-024-66923-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024] Open
Abstract
Cerebrospinal fluid (CSF) core biomarkers of Alzheimer's disease (AD), including amyloid peptide beta-42 (Aβ42), Aβ42/40 ratio, and phosphorylated tau (pTau), are precious tools for supporting AD diagnosis. However, their use in clinical practice is limited due to the invasiveness of CSF collection. Thus, there is intensive research to find alternative, noninvasive, and widely accessible biological matrices to measure AD core biomarkers. In this study, we measured AD core biomarkers in saliva and plasma by a fully automated platform. We enrolled all consecutive patients with cognitive decline. For each patient, we measured Aβ42, Aβ40, and pTau levels in CSF, saliva, and plasma by Lumipulse G1200 (Fujirebio). We included forty-two patients, of whom 27 had AD. Levels of all biomarkers significantly differed in the three biofluids, with saliva having the lowest and CSF the highest levels of Aβ42, Aβ40, and pTau. A positive correlation of pTau, Aβ42/40 ratio, and pTau/Aβ42 ratio levels in CSF and plasma was detected, while no correlation between any biomarker in CSF and saliva was found. Our findings suggest that plasma but not saliva could represent a surrogate biofluid for measuring core AD biomarkers. Specifically, plasma Aβ42/40 ratio, pTau/Aβ42 ratio, and pTau could serve as surrogates of the corresponding CSF biomarkers.
Collapse
Affiliation(s)
- Luisa Agnello
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Rosaria Vincenza Giglio
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
- Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| | - Fabio Del Ben
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO)-IRCCS, Aviano, Italy
| | - Tommaso Piccoli
- Neurology Unit, Department of Biomedicine, Neuroscience, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Tiziana Colletti
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Concetta Scazzone
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Bruna Lo Sasso
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
- Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| | - Anna Maria Ciaccio
- Internal Medicine and Medical Specialties "G. D'Alessandro", Department of Health Promotion, Maternal, and Infant Care, University of Palermo, Palermo, Italy
| | - Caterina Maria Gambino
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
- Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| | - Giuseppe Salemi
- Neurology Unit, Department of Biomedicine, Neuroscience, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Marcello Ciaccio
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences, and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy.
- Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy.
| |
Collapse
|
6
|
Nilsson J, Pichet Binette A, Palmqvist S, Brum WS, Janelidze S, Ashton NJ, Spotorno N, Stomrud E, Gobom J, Zetterberg H, Brinkmalm A, Blennow K, Hansson O. Cerebrospinal fluid biomarker panel for synaptic dysfunction in a broad spectrum of neurodegenerative diseases. Brain 2024; 147:2414-2427. [PMID: 38325331 PMCID: PMC11224614 DOI: 10.1093/brain/awae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/31/2023] [Accepted: 01/21/2024] [Indexed: 02/09/2024] Open
Abstract
Synaptic dysfunction and degeneration is likely the key pathophysiology for the progression of cognitive decline in various dementia disorders. Synaptic status can be monitored by measuring synaptic proteins in CSF. In this study, both known and new synaptic proteins were investigated and compared as potential biomarkers of synaptic dysfunction, particularly in the context of Alzheimer's disease (AD). Seventeen synaptic proteins were quantified in CSF using two different targeted mass spectrometry assays in the prospective Swedish BioFINDER-2 study. The study included 958 individuals, characterized as having mild cognitive impairment (MCI, n = 205), AD dementia (n = 149) and a spectrum of other neurodegenerative diseases (n = 171), in addition to cognitively unimpaired individuals (CU, n = 443). Synaptic protein levels were compared between diagnostic groups and their associations with cognitive decline and key neuroimaging measures (amyloid-β-PET, tau-PET and cortical thickness) were assessed. Among the 17 synaptic proteins examined, 14 were specifically elevated in the AD continuum. SNAP-25, 14-3-3 zeta/delta, β-synuclein, and neurogranin exhibited the highest discriminatory accuracy in differentiating AD dementia from controls (areas under the curve = 0.81-0.93). SNAP-25 and 14-3-3 zeta/delta also had the strongest associations with tau-PET, amyloid-β-PET and cortical thickness at baseline and were associated with longitudinal changes in these imaging biomarkers [β(standard error, SE) = -0.056(0.0006) to 0.058(0.005), P < 0.0001]. SNAP-25 was the strongest predictor of progression to AD dementia in non-demented individuals (hazard ratio = 2.11). In contrast, neuronal pentraxins were decreased in all neurodegenerative diseases (except for Parkinson's disease), and NPTX2 showed the strongest associations with subsequent cognitive decline [longitudinal Mini-Mental State Examination: β(SE) = 0.57(0.1), P ≤ 0.0001; and mPACC: β(SE) = 0.095(0.024), P ≤ 0.001] across the AD continuum. Interestingly, utilizing a ratio of the proteins that displayed higher levels in AD, such as SNAP-25 or 14-3-3 zeta/delta, over NPTX2 improved the biomarkers' associations with cognitive decline and brain atrophy. We found 14-3-3 zeta/delta and SNAP-25 to be especially promising as synaptic biomarkers of pathophysiological changes in AD. Neuronal pentraxins were identified as general indicators of neurodegeneration and associated with cognitive decline across various neurodegenerative dementias. Cognitive decline and brain atrophy were best predicted by ratios of SNAP-25/NPTX2 and 14-3-3 zeta/delta/NPTX2.
Collapse
Affiliation(s)
- Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, 4011 Stavanger, Norway
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London SE5 9RX, UK
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London SE5 8AF, UK
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
- Fluid Biomarker Laboratory, UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 30 Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, 75646 Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei 230036, P.R. China
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| |
Collapse
|
7
|
Warmenhoven N, Salvadó G, Janelidze S, Mattsson-Carlgren N, Bali D, Dolado AO, Kolb H, Triana-Baltzer G, Barthélemy NR, Schindler SE, Aschenbrenner AJ, Raji CA, Benzinger TL, Morris JC, Ibanez L, Timsina J, Cruchaga C, Bateman RJ, Ashton N, Arslan B, Zetterberg H, Blennow K, Pichet Binette A, Hansson O. A Comprehensive Head-to-Head Comparison of Key Plasma Phosphorylated Tau 217 Biomarker Tests. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.02.24309629. [PMID: 39006421 PMCID: PMC11245081 DOI: 10.1101/2024.07.02.24309629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Plasma phosphorylated-tau 217 (p-tau217) is currently the most promising biomarkers for reliable detection of Alzheimer's disease (AD) pathology. Various p-tau217 assays have been developed, but their relative performance is unclear. We compared key plasma p-tau217 tests using cross-sectional and longitudinal measures of amyloid-β (Aβ)-PET, tau-PET, and cognition as outcomes, and benchmarked them against cerebrospinal fluid (CSF) biomarker tests. Samples from 998 individuals (mean[range] age 68.5[20.0-92.5], 53% female) from the Swedish BioFINDER-2 cohort were analyzed. Plasma p-tau217 was measured with mass spectrometry (MS) assays (the ratio between phosphorylated and non-phosphorylated [%p-tau217WashU]and ptau217WashU) as well as with immunoassays (p-tau217Lilly, p-tau217Janssen, p-tau217ALZpath). CSF biomarkers included p-tau217Lilly, and the FDA-approved p-tau181/Aβ42Elecsys and p-tau181Elecsys. All plasma p-tau217 tests exhibited high ability to detect abnormal Aβ-PET (AUC range: 0.91-0.96) and tau-PET (AUC range: 0.94-0.97). Plasma %p-tau217WashU had the highest performance, with significantly higher AUCs than all the immunoassays (P diff<0.007). For detecting Aβ-PET status, %p-tau217WashU had an accuracy of 0.93 (immunoassays: 0.83-0.88), sensitivity of 91% (immunoassays: 84-87%), and a specificity of 94% (immunoassays: 85-89%). Among immunoassays, p-tau217Lilly and plasma p-tau217ALZpath had higher AUCs than plasma p-tau217Janssen for Aβ-PET status (P diff<0.006), and p-tau217Lilly outperformed plasma p-tau217ALZpath for tau-PET status (P diff=0.025). Plasma %p-tau217WashU exhibited higher associations with all PET load outcomes compared to immunoassays; baseline Aβ-PET load (R2: 0.72; immunoassays: 0.47-0.58; Pdiff<0.001), baseline tau-PET load (R2: 0.51; immunoassays: 0.38-0.45; Pdiff<0.001), longitudinal Aβ-PET load (R2: 0.53; immunoassays: 0.31-0.38; Pdiff<0.001) and longitudinal tau-PET load (R2: 0.50; immunoassays: 0.35-0.43; Pdiff<0.014). Among immunoassays, plasma p-tau217Lilly was more strongly associated with Aβ-PET load than plasma p-tau217Janssen (P diff<0.020) and with tau-PET load than both plasma p-tau217Janssen and plasma p-tau217ALZpath (all P diff<0.010). Plasma %p-tau217 also correlated more strongly with baseline cognition (Mini-Mental State Examination[MMSE]) than all immunoassays (R2 %p-tau217WashU: 0.33; immunoassays: 0.27-0.30; P diff<0.024). The main results were replicated in an external cohort from Washington University in St Louis (n =219). Finally, p-tau217Nulisa showed similar performance to other immunoassays in subsets of both cohorts. In summary, both MS- and immunoassay-based p-tau217 tests generally perform well in identifying Aβ-PET, tau-PET, and cognitive abnormalities, but %p-tau217WashU performed significantly better than all the examined immunoassays. Plasma %p-tau217 may be considered as a stand-alone confirmatory test for AD pathology, while some immunoassays might be better suited as triage tests where positive results are confirmed with a second test.
Collapse
Affiliation(s)
- Noëlle Warmenhoven
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Anna Orduña Dolado
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Johnson and Johnson Innovative Medicine, San Diego, CA, USA
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Johnson and Johnson Innovative Medicine, San Diego, CA, USA
| | - Nicolas R. Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Cyrus A. Raji
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University St. Louis, MO, USA
| | - Randall J. Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
8
|
Ho S, Darrow J, De Simone F, Calabro A, Gannon S, Esquivel R, Thakker P, Khingelova K, Rao A, Zhang Y, Moghekar A. Assessment of Preanalytical Cerebrospinal Fluid Handling and Storage Factors on Measurement of Aβ1-42, Aβ1-40, and pTau181 Using an Automated Chemiluminescent Platform. J Appl Lab Med 2024; 9:789-802. [PMID: 38712812 DOI: 10.1093/jalm/jfae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/11/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Standardizing cerebrospinal fluid (CSF) laboratory protocols will improve the reliability and availability of clinical biomarker testing required for prescription of novel Alzheimer disease (AD) therapies. This study evaluated several preanalytical handling and storage factors common to β-amyloid1-42 (Aβ1-42), β-amyloid1-40 (Aβ1-40), and phosphorylated tau (pTau181) concentrations including storage at different temperatures, extended cap contact, various mixing methods, and multiple freeze-thaw cycles. METHODS Aβ1-42, Aβ1-40, and pTau181 concentrations were measured using LUMIPULSE G1200 automated assays. Samples were collected in polypropylene tubes of various volumes. Sample cap-contact was evaluated by storing samples in upright and inverted positions at either 4°C for 1 week or -80°C for 1 month. To assess mixing methods, samples were freeze-thawed and mixed by inversion, vortex, horizontal roller, or unmixed prior to assay sampling. The impact of successive freeze-thaw cycles was assessed through freezing, thawing, and analyzing CSF samples. RESULTS Short-term storage at 4°C did not affect Aβ1-42, Aβ1-40, or pTau181 measurements in any tube type. Tube cap contact affected Aβ1-42 in 2.5 mL tubes and pTau181 levels in 10 mL tubes. No difference was observed between mixing methods. After 4 freeze-thaw cycles, Aβ1-42 significantly decreased but Aβ1-40 remained unchanged. Utilizing the Aβ1-42/Aβ1-40 ratio, Aβ1-42 values normalized, maintaining ratio values within ±5% of baseline measurements. CONCLUSIONS Storage of CSF at 4°C for 1 week or -80°C for 1 month did not significantly affect Aβ1-42, Aβ1-40, pTau181, or associated ratio measurements. Tube cap-contact impacted pTau181 and pTau181/Aβ1-42 values in larger tubes. Mixing methods are equivalent. The Aβ1-42/Aβ1-40 ratio compensates for freeze-thaw variability up to 4 cycles.
Collapse
Affiliation(s)
- Sara Ho
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Jacqueline Darrow
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | | | | | - Sara Gannon
- Fujirebio Diagnostics Inc., Malvern, PA, United States
| | | | - Parmi Thakker
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Kristina Khingelova
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Aruna Rao
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Yifan Zhang
- Department of Biostatistics, Johns Hopkins School of Public Health, Baltimore, MD, United States
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Schneider TR, Stöckli L, Felbecker A, Nirmalraj PN. Protein fibril aggregation on red blood cells: a potential biomarker to distinguish neurodegenerative diseases from healthy aging. Brain Commun 2024; 6:fcae180. [PMID: 38873003 PMCID: PMC11170662 DOI: 10.1093/braincomms/fcae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 04/19/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024] Open
Abstract
Neurodegenerative diseases like Alzheimer's disease are characterized by the accumulation of misfolded proteins into fibrils in the brain. Atomic force microscopy is a nanoscale imaging technique that can be used to resolve and quantify protein aggregates from oligomers to fibrils. Recently, we characterized protein fibrillar aggregates adsorbed on the surface of red blood cells with atomic force microscopy from patients with neurocognitive disorders, suggesting a novel Alzheimer's disease biomarker. However, the age association of fibril deposits on red blood cells has not yet been studied in detail in healthy adults. Here, we used atomic force microscopy to visualize and quantify fibril coverage on red blood cells in 50 healthy adults and 37 memory clinic patients. Fibrillar protein deposits sporadically appeared in healthy individuals but were much more prevalent in patients with neurodegenerative disease, especially those with Alzheimer's disease as confirmed by positive CSF amyloid beta 1-42/1-40 ratios. The prevalence of fibrils on the red blood cell surface did not significantly correlate with age in either healthy individuals or Alzheimer's disease patients. The overlap in fibril prevalence on red blood cells between Alzheimer's disease and amyloid-negative patients suggests that fibril deposition on red blood cells could occur in various neurodegenerative diseases. Quantifying red blood cell protein fibril morphology and prevalence on red blood cells could serve as a sensitive biomarker for neurodegeneration, distinguishing between healthy individuals and those with neurodegenerative diseases. Future studies that combine atomic force microscopy with immunofluorescence techniques in larger-scale studies could further identify the chemical nature of these fibrils, paving the way for a comprehensive, non-invasive biomarker platform for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Luisa Stöckli
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| | - Ansgar Felbecker
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| | - Peter Niraj Nirmalraj
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf CH-8600, Switzerland
| |
Collapse
|
10
|
Wuestefeld A, Binette AP, van Westen D, Strandberg O, Stomrud E, Mattsson-Carlgren N, Janelidze S, Smith R, Palmqvist S, Baumeister H, Berron D, Yushkevich PA, Hansson O, Spotorno N, Wisse LEM. Medial temporal lobe atrophy patterns in early- versus late-onset amnestic Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.594976. [PMID: 38826333 PMCID: PMC11142072 DOI: 10.1101/2024.05.21.594976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background The medial temporal lobe (MTL) is hypothesized to be relatively spared in early-onset Alzheimer's disease (EOAD). Yet, detailed examination of MTL subfield volumes and drivers of atrophy in amnestic EOAD is lacking. Methods BioFINDER-2 participants with memory impairment, abnormal amyloid-β status and tau-PET were included. Forty-one EOAD individuals aged ≤65 years and, as comparison, late-onset AD (LOAD, ≥70 years, n=154) and Aβ-negative cognitively unimpaired controls were included. MTL subregions and biomarkers of (co-)pathologies were measured. Results AD groups showed smaller MTL subregions compared to controls. Atrophy patterns were similar across AD groups, although LOAD showed thinner entorhinal cortices compared to EOAD. EOAD showed lower WMH compared to LOAD. No differences in MTL tau-PET or transactive response DNA binding protein 43-proxy positivity was found. Conclusions We found in vivo evidence for MTL atrophy in amnestic EOAD and overall similar levels to LOAD of MTL tau pathology and co-pathologies.
Collapse
Affiliation(s)
- Anika Wuestefeld
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
| | - Danielle van Westen
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, 22242 Lund, Sweden
- Image and Function, Skåne University Hospital, 22242 Lund Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 20502 Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
- Department of Neurology, Skåne University Hospital, 22242 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 20502 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 20502 Malmö, Sweden
| | - Hannah Baumeister
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Paul A. Yushkevich
- Penn Image Computing and Science Laboratory (PICSL), Department of Radiology, University of Pennsylvania, Philadelphia 19104, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 20502 Malmö, Sweden
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 22242 Lund, Sweden
| | - Laura EM Wisse
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, 22242 Lund, Sweden
| |
Collapse
|
11
|
Algeciras‐Schimnich A, Bornhorst JA. Importance of cerebrospinal fluid (CSF) collection protocol for the accurate diagnosis of Alzheimer's disease when using CSF biomarkers. Alzheimers Dement 2024; 20:3657-3658. [PMID: 38288880 PMCID: PMC11095420 DOI: 10.1002/alz.13721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 05/16/2024]
Affiliation(s)
| | - Joshua A. Bornhorst
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
12
|
Barthélemy NR, Salvadó G, Schindler SE, He Y, Janelidze S, Collij LE, Saef B, Henson RL, Chen CD, Gordon BA, Li Y, La Joie R, Benzinger TLS, Morris JC, Mattsson-Carlgren N, Palmqvist S, Ossenkoppele R, Rabinovici GD, Stomrud E, Bateman RJ, Hansson O. Highly accurate blood test for Alzheimer's disease is similar or superior to clinical cerebrospinal fluid tests. Nat Med 2024; 30:1085-1095. [PMID: 38382645 PMCID: PMC11031399 DOI: 10.1038/s41591-024-02869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/12/2024] [Indexed: 02/23/2024]
Abstract
With the emergence of Alzheimer's disease (AD) disease-modifying therapies, identifying patients who could benefit from these treatments becomes critical. In this study, we evaluated whether a precise blood test could perform as well as established cerebrospinal fluid (CSF) tests in detecting amyloid-β (Aβ) plaques and tau tangles. Plasma %p-tau217 (ratio of phosporylated-tau217 to non-phosphorylated tau) was analyzed by mass spectrometry in the Swedish BioFINDER-2 cohort (n = 1,422) and the US Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC) cohort (n = 337). Matched CSF samples were analyzed with clinically used and FDA-approved automated immunoassays for Aβ42/40 and p-tau181/Aβ42. The primary and secondary outcomes were detection of brain Aβ or tau pathology, respectively, using positron emission tomography (PET) imaging as the reference standard. Main analyses were focused on individuals with cognitive impairment (mild cognitive impairment and mild dementia), which is the target population for available disease-modifying treatments. Plasma %p-tau217 was clinically equivalent to FDA-approved CSF tests in classifying Aβ PET status, with an area under the curve (AUC) for both between 0.95 and 0.97. Plasma %p-tau217 was generally superior to CSF tests in classification of tau-PET with AUCs of 0.95-0.98. In cognitively impaired subcohorts (BioFINDER-2: n = 720; Knight ADRC: n = 50), plasma %p-tau217 had an accuracy, a positive predictive value and a negative predictive value of 89-90% for Aβ PET and 87-88% for tau PET status, which was clinically equivalent to CSF tests, further improving to 95% using a two-cutoffs approach. Blood plasma %p-tau217 demonstrated performance that was clinically equivalent or superior to clinically used FDA-approved CSF tests in the detection of AD pathology. Use of high-performance blood tests in clinical practice can improve access to accurate AD diagnosis and AD-specific treatments.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lyduine E Collij
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Benjamin Saef
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel L Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Gil D Rabinovici
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
13
|
Patel KJ, Yang D, Feldman HH, Hsiung GR, Nygaard HB, Best JR, Dwosh E, Robillard JM, DeMarco ML. Personal value of Alzheimer's disease biomarker testing and result disclosure from the patient and care partner perspective. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12463. [PMID: 38596482 PMCID: PMC10999946 DOI: 10.1002/trc2.12463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/19/2023] [Accepted: 01/12/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION We described patients' and care partners' experiences with Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarker testing and result disclosure in routine care. METHODS IMPACT-AD BC is an observational study of clinic patients who underwent AD CSF biomarker testing as part of their routine medical care (n = 142). In the personal utility arm of the study, semi-structured phone interviews were conducted with a subset of patients (n = 34), and separately with their care partners (n = 31). Post-disclosure interviews were conducted ∼1 month and ∼6 months after biomarker result disclosure and investigated the patients' decision-making process around testing, impact of receiving results, wellness and lifestyle changes, and future planning. RESULTS A majority of patients (90%) rated their decision to undergo testing as "easy." Post-disclosure, the majority (82%) reported overall positive feelings from having greater certainty and the ability to plan ahead, and results spurred them to adopt/continue healthy behaviors such as exercise (84%) and cognitive activities (54%). Care partners expressed relief from having more diagnostic certainty, increased appreciation of future caregiving responsibilities, and a desire to connect with support resources. DISCUSSION Perspectives of persons with lived experience in dementia provide new insight into the value of biomarker testing and should be included as part of evidence-guided considerations for pre-test counseling and result disclosure. Moreover, study findings identify an interval when patients and care partners are highly receptive to positive lifestyle and medical interventions.
Collapse
Affiliation(s)
- Khushbu J. Patel
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverCanada
| | - David Yang
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverCanada
| | - Howard H. Feldman
- Department of NeurosciencesUniversity of California San DiegoSan DiegoCaliforniaUSA
- Alzheimer Disease Cooperative Study, University of California San DiegoSan DiegoCaliforniaUSA
- Alzheimer's and Related Neurodegenerative Research, University of California San DiegoSan DiegoCaliforniaUSA
| | - Ging‐Yuek R. Hsiung
- Division of NeurologyDepartment of MedicineUniversity of British ColumbiaVancouverCanada
- Djavad Mowafaghian Centre for Brain HealthDepartment of MedicineUniversity of British ColumbiaVancouverCanada
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders, University of British ColumbiaVancouverCanada
| | - Haakon B. Nygaard
- Division of NeurologyDepartment of MedicineUniversity of British ColumbiaVancouverCanada
- Djavad Mowafaghian Centre for Brain HealthDepartment of MedicineUniversity of British ColumbiaVancouverCanada
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders, University of British ColumbiaVancouverCanada
| | - John R. Best
- Gerontology Research Centre, Simon Fraser UniversityVancouverCanada
| | - Emily Dwosh
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders, University of British ColumbiaVancouverCanada
- Department of Medical GeneticsUniversity of British ColumbiaVancouverCanada
| | - Julie M. Robillard
- Division of NeurologyDepartment of MedicineUniversity of British ColumbiaVancouverCanada
- Djavad Mowafaghian Centre for Brain HealthDepartment of MedicineUniversity of British ColumbiaVancouverCanada
| | - Mari L. DeMarco
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverCanada
- Department of Pathology and Laboratory MedicineSt. Paul's Hospital, Providence Health CareVancouverCanada
| |
Collapse
|
14
|
Jonaitis EM, Jeffers B, VandenLangenberg M, Ma Y, Van Hulle C, Langhough R, Du L, Chin NA, Przybelski RJ, Hogan KJ, Christian BT, Betthauser TJ, Okonkwo OC, Bendlin BB, Asthana S, Carlsson CM, Johnson SC. CSF Biomarkers in Longitudinal Alzheimer Disease Cohorts: Pre-Analytic Challenges. Clin Chem 2024; 70:538-550. [PMID: 38431278 PMCID: PMC10908554 DOI: 10.1093/clinchem/hvad221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/27/2023] [Indexed: 03/05/2024]
Abstract
BACKGROUND The sensitivity of amyloid to pre-analytic factors complicates cerebrospinal fluid (CSF) diagnostics for Alzheimer disease. We report reliability and validity evidence for automated immunoassays from frozen and fresh CSF samples in an ongoing, single-site research program. METHODS CSF samples were obtained from 2 Wisconsin cohorts (1256 measurements; 727 participants). Levels of amyloid beta 1-42 (Aβ42), phosphorylated tau 181 (pTau181), and total tau (tTau) were obtained using an Elecsys cobas e 601 platform. Repeatability and fixed effects of storage tube type, extraction method, and freezing were assessed via mixed models. Concordance with amyloid positron emission tomography (PET) was investigated with 238 participants having a temporally proximal PET scan. RESULTS Repeatability was high with intraclass correlation (ICC) ≥0.9, but tube type strongly affected measurements. Discriminative accuracy for PET amyloid positivity was strong across tube types (area under the curve [AUC]: Aβ42, 0.87; pTau181Aβ42 , 0.96), although optimal thresholds differed. CONCLUSIONS Under real-world conditions, the Elecsys platform had high repeatability. However, strong effects of pre-analytic factors suggest caution in drawing longitudinal inferences.
Collapse
Affiliation(s)
- Erin M Jonaitis
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Beckie Jeffers
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Monica VandenLangenberg
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Yue Ma
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Carol Van Hulle
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Rebecca Langhough
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Lianlian Du
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Nathaniel A Chin
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Robert J Przybelski
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Kirk J Hogan
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Bradley T Christian
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Psychiatry, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Tobey J Betthauser
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Barbara B Bendlin
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Sanjay Asthana
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Cynthia M Carlsson
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Sterling C Johnson
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| |
Collapse
|
15
|
Yu S, Zou Y, Ma X, Wang D, Luo W, Tang Y, Mu D, Zhang R, Cheng X, Qiu L. Evolution of LC-MS/MS in clinical laboratories. Clin Chim Acta 2024; 555:117797. [PMID: 38280490 DOI: 10.1016/j.cca.2024.117797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/29/2024]
Abstract
Liquid chromatography-tandem mass spectrometry (LC-MS/MS) has attracted significant attention in clinical practice owing to its numerous advantages. However, the widespread adoption of this technique is hindered by certain limitations, such as inappropriate analyte selection, low levels of automation, and a lack of specific reference intervals and quality control programs. This review comprehensively summarizes the current challenges associated with LC-MS/MS and proposes potential resolutions. The principle of utility should guide the selection of biomarkers, prioritizing their practical value over sheer quantity. To achieve full-process automation, methodological innovation is crucial for developing high-throughput equipment. Establishing reference intervals for mass spectrometry-based assays across multiple centers and diverse populations is essential for accurate result interpretation. Additionally, the development of commercial quality control materials assumes pivotal importance in ensuring assay reliability and reproducibility. Harmonization and standardization efforts should focus on the development of reference methods and materials for the clinical use of LC-MS/MS. In the future, commercial assay kits and laboratory-developed tests (LDTs) are expected to coexist in clinical laboratories, each offering distinct advantages. The collaborative efforts of diverse professionals is vital for addressing the challenges associated with the clinical application of LC-MS/MS. The anticipated advancements include simplification, increased automation, intelligence, and the standardization of LC-MS/MS, ultimately facilitating its seamless integration into clinical routines for both technicians and clinicians.
Collapse
Affiliation(s)
- Songlin Yu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Xiaoli Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Danchen Wang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Wei Luo
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Yueming Tang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Danni Mu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Ruiping Zhang
- Department of Laboratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xinqi Cheng
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China.
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China.
| |
Collapse
|
16
|
Moonen JEF, Haan R, Bos I, Teunissen C, van de Giessen E, Tomassen J, den Braber A, van der Landen SM, de Geus EJC, Legdeur N, van Harten AC, Trieu C, de Boer C, Kroeze L, Barkhof F, Visser PJ, van der Flier WM. Contributions of amyloid beta and cerebral small vessel disease in clinical decline. Alzheimers Dement 2024; 20:1868-1880. [PMID: 38146222 PMCID: PMC10984432 DOI: 10.1002/alz.13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION We assessed whether co-morbid small vessel disease (SVD) has clinical predictive value in preclinical or prodromal Alzheimer's disease. METHODS In 1090 non-demented participants (65.4 ± 10.7 years) SVD was assessed with magnetic resonance imaging and amyloid beta (Aβ) with lumbar puncture and/or positron emission tomography scan (mean follow-up for cognitive function 3.1 ± 2.4 years). RESULTS Thirty-nine percent had neither Aβ nor SVD (A-V-), 21% had SVD only (A-V+), 23% Aβ only (A+V-), and 17% had both (A+V+). Pooled cohort linear mixed model analyses demonstrated that compared to A-V- (reference), A+V- had a faster rate of cognitive decline. Co-morbid SVD (A+V+) did not further increase rate of decline. Cox regression showed that dementia risk was modestly increased in A-V+ (hazard ratio [95% confidence interval: 1.8 [1.0-3.2]) and most strongly in A+ groups. Also, mortality risk was increased in A+ groups. DISCUSSION In non-demented persons Aβ was predictive of cognitive decline, dementia, and mortality. SVD modestly predicts dementia in A-, but did not increase deleterious effects in A+. HIGHLIGHTS Amyloid beta (Aβ; A) was predictive for cognitive decline, dementia, and mortality. Small vessel disease (SVD) had no additional deleterious effects in A+. SVD modestly predicted dementia in A-. Aβ should be assessed even when magnetic resonance imaging indicates vascular cognitive impairment.
Collapse
Affiliation(s)
- Justine E. F. Moonen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Renée Haan
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Isabelle Bos
- Nivel, Research Institute for Better CareUtrechtthe Netherlands
| | - Charlotte Teunissen
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Sophie M. van der Landen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Eco J. C. de Geus
- Department of Biological PsychologyVU UniversityAmsterdamthe Netherlands
| | - Nienke Legdeur
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Argonde C. van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Calvin Trieu
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Casper de Boer
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Lior Kroeze
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Institute of Healthcare Engineering and the Institute of Neurology, University College LondonLondonUK
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNS), Maastricht UniversityMaastrichtthe Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of NeurogeriatricsKarolinska InstitutetSolnaSweden
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| |
Collapse
|
17
|
Igeta Y, Hemmi I, Yuyama K, Ouchi Y. Odor identification score as an alternative method for early identification of amyloidogenesis in Alzheimer's disease. Sci Rep 2024; 14:4658. [PMID: 38409432 PMCID: PMC10897211 DOI: 10.1038/s41598-024-54322-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 02/11/2024] [Indexed: 02/28/2024] Open
Abstract
A simple screening test to identify the early stages of Alzheimer's disease (AD) is urgently needed. We investigated whether odor identification impairment can be used to differentiate between stages of the A/T/N classification (amyloid, tau, neurodegeneration) in individuals with amnestic mild cognitive impairment or AD and in healthy controls. We collected data from 132 Japanese participants visiting the Toranomon Hospital dementia outpatient clinic. The odor identification scores correlated significantly with major neuropsychological scores, regardless of apolipoprotein E4 status, and with effective cerebrospinal fluid (CSF) biomarkers [amyloid β 42 (Aβ42) and the Aβ42/40 and phosphorylated Tau (p-Tau)/Aβ42 ratios] but not with ineffective biomarkers [Aβ40 and the p-Tau/total Tau ratio]. A weak positive correlation was observed between the corrected odor identification score (adjusted for age, sex, ApoE4 and MMSE), CSF Aβ42, and the Aβ42/40 ratio. The odor identification score demonstrated excellent discriminative power for the amyloidogenesis stage , according to the A/T/N classification, but was unsuitable for differentiating between the p-Tau accumulation and the neurodegeneration stages. After twelve odor species were analyzed, a version of the score comprising only four odors-India ink, wood, curry, and sweaty socks-proved highly effective in identifying AD amyloidogenesis, showing promise for the screening of preclinical AD.
Collapse
Affiliation(s)
- Yukifusa Igeta
- Department of Dementia, Dementia Center, Federation of National Public Service Personnel Mutual Aid Associations, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan.
- Division of Dementia Research, Okinaka Memorial Institute for Medical Research, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan.
| | - Isao Hemmi
- Japanese Red Cross College of Nursing, 4-1-3 Hiroo, Shibuya-ku, Tokyo, 150-0012, Japan
| | - Kohei Yuyama
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, 001-0021, Japan
| | - Yasuyoshi Ouchi
- Department of Dementia, Dementia Center, Federation of National Public Service Personnel Mutual Aid Associations, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
- Division of Dementia Research, Okinaka Memorial Institute for Medical Research, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| |
Collapse
|
18
|
Yuyama K, Sun H, Fujii R, Hemmi I, Ueda K, Igeta Y. Extracellular vesicle proteome unveils cathepsin B connection to Alzheimer's disease pathogenesis. Brain 2024; 147:627-636. [PMID: 38071653 PMCID: PMC10834236 DOI: 10.1093/brain/awad361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 02/03/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles that are released extracellularly and considered to be implicated in the pathogenesis of neurodegenerative diseases including Alzheimer's disease. Here, CSF EVs of 16 ATN-classified cases were subjected to quantitative proteome analysis. In these CSF EVs, levels of 11 proteins were significantly altered during the ATN stage transitions (P < 0.05 and fold-change > 2.0). These proteins were thought to be associated with Alzheimer's disease pathogenesis and represent candidate biomarkers for pathogenic stage classification. Enzyme-linked immunosorbent assay analysis of CSF and plasma EVs revealed altered levels of cathepsin B (CatB) during the ATN transition (seven ATN groups in validation set, n = 136). The CSF and plasma EV CatB levels showed a negative correlation with CSF amyloid-β42 concentrations. This proteomic landscape of CSF EVs in ATN classifications can depict the molecular framework of Alzheimer's disease progression, and CatB may be considered a promising candidate biomarker and therapeutic target in Alzheimer's disease amyloid pathology.
Collapse
Affiliation(s)
- Kohei Yuyama
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Hui Sun
- Lipid Biofunction Section, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Risa Fujii
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Isao Hemmi
- Department of Nursing, Japanese Red Cross College of Nursing, Tokyo 150-0012, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 035-8550, Japan
| | - Yukifusa Igeta
- Department of Dementia, Dementia Center, Toranomon Hospital, Tokyo 105-8470, Japan
- Division of Dementia Research, Okinaka Memorial Institute for Medical Research, Tokyo 105-8470, Japan
| |
Collapse
|
19
|
Giuffrè GM, Quaranta D, Citro S, Morganti TG, Martellacci N, Vita MG, Rossini PM, Calabresi P, Marra C. Associations Between Free and Cued Selective Reminding Test and Cerebrospinal Fluid Biomarkers in Amnestic Mild Cognitive Impairment. J Alzheimers Dis 2024; 100:713-723. [PMID: 38905044 DOI: 10.3233/jad-240150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
Background The Free and Cued Selective Reminding Test (FCSRT), assessing verbal episodic memory with controlled learning and semantic cueing, has been recommended for detecting the genuine encoding and storage deficits characterizing AD-related memory disorders. Objective The present study aims at investigating the ability of FCSRT in predicting cerebrospinal fluid (CSF) evidence of amyloid-β positivity in subjects with amnestic mild cognitive impairment (aMCI) and exploring its associations with amyloidopathy, tauopathy and neurodegeneration biomarkers. Methods 120 aMCI subjects underwent comprehensive neurological and neuropsychological examinations, including the FCSRT assessment, and CSF collection; CSF Aβ42/40 ratio, p-tau181, and total-tau quantification were conducted by an automated CLEIA method on Lumipulse G1200. Based on the Aβ42/40 ratio value, subjects were classified as either A+ or A-. Results All FCSRT subitem scores were significantly lower in A+ group and significantly predicted the amyloid-β status, with Immediate Total Recall (ITR) being the best predictor. No significant correlations were found between FCSRT and CSF biomarkers in the A- aMCI group, while in the A+ aMCI group, all FCSRT subitem scores were negatively correlated with CSF p-tau181 and total-tau, but not with the Aβ42/40 ratio. Conclusions FCSRT confirms its validity as a tool for the diagnosis of AD, being able to predict the presence of amyloid-β deposition with high specificity. The associations between FCSRT subitem scores and CSF p-tau-181 and total-tau levels in aMCI due to AD could further encourage the clinical use of this simple and cost-effective test in the evaluation of individuals with aMCI.
Collapse
Affiliation(s)
- Guido Maria Giuffrè
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Davide Quaranta
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Salvatore Citro
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Tommaso Giuseppe Morganti
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Noemi Martellacci
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Gabriella Vita
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paolo Maria Rossini
- Department of Neuroscience and Neurorehabilitation, Brain Connectivity Laboratory, IRCCS San Raffaele Roma, Rome, Italy
| | - Paolo Calabresi
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Camillo Marra
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
20
|
Sánchez‐Soblechero A, Berbel A, Villarejo A, Palmí‐Cortés I, Vieira A, Gil‐Moreno MJ, Fernández C, Martín‐Montes Ã, Carreras MT, Fernández Y, Puertas C, Blanco‐Palmero V, Llamas S, González‐Sánchez M, Lapeña T, de Luis P, Manzano S, Olazarán J. Translating NIA-AA criteria into usual practice: Report from the ReDeMa Project. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12451. [PMID: 38505833 PMCID: PMC10948948 DOI: 10.1002/trc2.12451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/23/2023] [Accepted: 12/30/2023] [Indexed: 03/21/2024]
Abstract
INTRODUCTION Biomarker-informed criteria were proposed for the diagnosis of Alzheimer's disease (AD) by the National Institute on Aging and the Alzheimer's Association (NIA-AA) in 2011; however, the adequacy of this criteria has not been sufficiently evaluated. METHODS ReDeMa (Red de Demencias de Madrid) is a regional cohort of patients attending memory and neurology clinics. Core cerebrospinal fluid biomarkers were obtained, NIA-AA diagnostic criteria were considered, and changes in diagnosis and management were evaluated. RESULTS A total of 233 patients were analyzed (mean age 70 years, 50% women, 73% AD). The diagnostic language was modified significantly, with a majority assumption of NIA-AA definitions (69%). Confidence in diagnosis increased from 70% to 92% (p < 0.0005) and management was changed in 71% of patient/caregivers. The influence of neurologist's age or expertise on study results was minimal. DISCUSSION The NIA-AA criteria are adequate and utile for usual practice in memory and neurology clinics, improving diagnostic confidence and significantly modifying patient management. HIGHLIGHTS Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers increase diagnostic certainty regardless of the neurologist.AD CSF biomarkers lead to changes in disease management .Biomarker-enriched, 2011 NIA-AA diagnostic criteria are adequate for usual practice.
Collapse
Affiliation(s)
| | | | | | - Itziar Palmí‐Cortés
- Neurology ServiceUniversity Hospital Infanta Sofía, San Sebastián de los ReyesMadridSpain
| | - Alba Vieira
- Neurology ServiceUniversity Hospital la PrincesaMadridSpain
| | | | | | - Ãngel Martín‐Montes
- Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital – Universidad Autónoma de Madrid)MadridSpain
| | | | - Yolanda Fernández
- Memory Disorders Clinic ‐ HM Hospitals and Neurology Service ‐ University Hospital Gregorio MarañónMadridSpain
| | - Carolina Puertas
- Clinical Biochemistry ServiceUniversity Hospital Gregorio MarañónMadridSpain
| | | | - Sara Llamas
- Neurology ServiceUniversity Hospital 12 de OctubreMadridSpain
| | - Marta González‐Sánchez
- Neurology ServiceUniversity Hospital 12 de OctubreMadridSpain
- Group of Neurodegenerative DiseasesUniversity Hospital 12 de Octubre Research Institute (imas12), and Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | | | | | | | - Javier Olazarán
- Memory Disorders Clinic ‐ HM HospitalsNeurology Service ‐ University Hospital Gregorio Marañón, and Maria Wolff FoundationMadridSpain
| |
Collapse
|
21
|
Kurihara M, Kondo S, Ohse K, Nojima H, Kikkawa-Saito E, Iwata A. Relationship Between Cerebrospinal Fluid Alzheimer's Disease Biomarker Values Measured via Lumipulse Assays and Conventional ELISA: Single-Center Experience and Systematic Review. J Alzheimers Dis 2024; 99:1077-1092. [PMID: 38759016 PMCID: PMC11191528 DOI: 10.3233/jad-240185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2024] [Indexed: 05/19/2024]
Abstract
Background Although Lumipulse assays and conventional ELISA are strongly correlated, the precise relationship between their measured values remains undetermined. Objective To determine the relationship between Lumipulse and ELISA measurement values. Methods Patients who underwent cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarker measurements and consented to biobanking between December 2021 and June 2023 were included. The relationship between values measured via Lumipulse assays and conventional ELISA were evaluated by Passing-Bablok analyses for amyloid-β 1-42 (Aβ42), total tau (t-tau), and phospho-tau 181 (p-tau 181). Studies using both assays were systematically searched for in PubMed and summarized after quality assessment. Results Regression line slopes and intercepts were 1.41 (1.23 to 1.60) and -77.8 (-198.4 to 44.5) for Aβ42, 0.94 (0.88 to 1.01) and 98.2 (76.9 to 114.4) for t-tau, and 1.60 (1.43 to 1.75) and -21.1 (-26.9 to -15.6) for p-tau181. Spearman's correlation coefficients were 0.90, 0.95, and 0.95 for Aβ42, t-tau, and p-tau181, respectively. We identified 13 other studies that included 2,117 patients in total. Aβ42 slope varied among studies, suggesting inter-lab difference of ELISA. The slope and intercept of t-tau were approximately 1 and 0, respectively, suggesting small proportional and systematic differences. Conversely, the p-tau181 slope was significantly higher than 1, distributed between 1.5-2 in most studies, with intercepts significantly lower than 0, suggesting proportional and systematic differences. Conclusions We characterized different relationship between measurement values for each biomarker, which may be useful for understanding the differences in CSF biomarker measurement values on different platforms and for future global harmonization.
Collapse
Affiliation(s)
- Masanori Kurihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
- Integrated Research Initiative for Living Well with Dementia, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Soichiro Kondo
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Kensuke Ohse
- Integrated Research Initiative for Living Well with Dementia, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | | | | | - Atsushi Iwata
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
- Integrated Research Initiative for Living Well with Dementia, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
22
|
Amato LG, Vergani AA, Lassi M, Fabbiani C, Mazzeo S, Burali R, Nacmias B, Sorbi S, Mannella R, Grippo A, Bessi V, Mazzoni A. Personalized modeling of Alzheimer's disease progression estimates neurodegeneration severity from EEG recordings. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12526. [PMID: 38371358 PMCID: PMC10870085 DOI: 10.1002/dad2.12526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Early identification of Alzheimer's disease (AD) is necessary for a timely onset of therapeutic care. However, cortical structural alterations associated with AD are difficult to discern. METHODS We developed a cortical model of AD-related neurodegeneration accounting for slowing of local dynamics and global connectivity degradation. In a monocentric study we collected electroencephalography (EEG) recordings at rest from participants in healthy (HC, n = 17), subjective cognitive decline (SCD, n = 58), and mild cognitive impairment (MCI, n = 44) conditions. For each patient, we estimated neurodegeneration model parameters based on individual EEG recordings. RESULTS Our model outperformed standard EEG analysis not only in discriminating between HC and MCI conditions (F1 score 0.95 vs 0.75) but also in identifying SCD patients with biological hallmarks of AD in the cerebrospinal fluid (recall 0.87 vs 0.50). DISCUSSION Personalized models could (1) support classification of MCI, (2) assess the presence of AD pathology, and (3) estimate the risk of cognitive decline progression, based only on economical and non-invasive EEG recordings. Highlights Personalized cortical model estimating structural alterations from EEG recordings.Discrimination of Mild Cognitive Impairment (MCI) and Healthy (HC) subjects (95%)Prediction of biological markers of Alzheimer's in Subjective Decline (SCD) Subjects (87%)Transition correctly predicted for 3/3 subjects that converted from SCD to MCI after 1y.
Collapse
Affiliation(s)
- Lorenzo Gaetano Amato
- The BioRobotics InstituteSant'Anna School of Advanced StudiesPisaItaly
- Department of Excellence in Robotics and AISant'Anna School of Advanced StudiesPisaItaly
| | - Alberto Arturo Vergani
- The BioRobotics InstituteSant'Anna School of Advanced StudiesPisaItaly
- Department of Excellence in Robotics and AISant'Anna School of Advanced StudiesPisaItaly
| | - Michael Lassi
- The BioRobotics InstituteSant'Anna School of Advanced StudiesPisaItaly
- Department of Excellence in Robotics and AISant'Anna School of Advanced StudiesPisaItaly
| | - Carlo Fabbiani
- IRCSS Fondazione Don Carlo GnocchiFlorenceItaly
- Department of NeurosciencePsychology, Drug Research and Child HealthCareggi University HospitalFlorenceItaly
| | - Salvatore Mazzeo
- IRCSS Fondazione Don Carlo GnocchiFlorenceItaly
- Department of NeurosciencePsychology, Drug Research and Child HealthCareggi University HospitalFlorenceItaly
| | | | - Benedetta Nacmias
- IRCSS Fondazione Don Carlo GnocchiFlorenceItaly
- Department of NeurosciencePsychology, Drug Research and Child HealthCareggi University HospitalFlorenceItaly
| | - Sandro Sorbi
- IRCSS Fondazione Don Carlo GnocchiFlorenceItaly
- Department of NeurosciencePsychology, Drug Research and Child HealthCareggi University HospitalFlorenceItaly
| | | | | | - Valentina Bessi
- Department of NeurosciencePsychology, Drug Research and Child HealthCareggi University HospitalFlorenceItaly
| | - Alberto Mazzoni
- The BioRobotics InstituteSant'Anna School of Advanced StudiesPisaItaly
- Department of Excellence in Robotics and AISant'Anna School of Advanced StudiesPisaItaly
| |
Collapse
|
23
|
Jethwa A, Stöckl L. Optimized Pre-analytical Handling Protocol for Blood-Based Biomarkers of Alzheimer's Disease. Methods Mol Biol 2024; 2785:67-73. [PMID: 38427188 DOI: 10.1007/978-1-0716-3774-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The therapeutic management of patients with Alzheimer's disease (AD) has been hindered by poor diagnostic accuracy. As such, there is an unmet clinical need for tools that can detect and diagnose the disease in its early stages. Compared with cerebrospinal fluid (CSF)-based biomarkers or positron emission tomography (PET), the use of reliable blood-based biomarkers could offer an accessible and minimally invasive method of streamlining diagnosis in the clinical setting. However, the influence of pre-analytical processing and sample handling parameters on the accurate measurement of protein biomarkers is well established, especially for AD CSF-based biomarkers. In this chapter, we provide recommendations for an optimal sample handling protocol for the analysis of blood-based biomarkers specifically for amyloid pathology in AD.
Collapse
|
24
|
Lehmann S, Schraen-Maschke S, Vidal JS, Blanc F, Paquet C, Allinquant B, Bombois S, Gabelle A, Delaby C, Hanon O. Blood Neurofilament Levels Predict Cognitive Decline across the Alzheimer's Disease Continuum. Int J Mol Sci 2023; 24:17361. [PMID: 38139190 PMCID: PMC10743700 DOI: 10.3390/ijms242417361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Neurofilament light chain (NfL) is a potential diagnostic and prognostic plasma biomarker for numerous neurological diseases including Alzheimer's disease (AD). In this study, we investigated the relationship between baseline plasma concentration of Nfl and Mild Cognitive Impairment in participants who did and did not have a clinically determined diagnosis of dementia by the end of the three-year study. Additionally, we explored the connection between baseline plasma concentration of NfL and AD dementia patients, considering their demographics, clinical features, and cognitive profiles. A total of 350 participants from the Biomarker of AmyLoid pepTide and AlZheimer's diseAse Risk (BALTAZAR) multicenter prospective study were investigated: 161 AD dementia participants and 189 MCI participants (of which 141 had amnestic MCI and 48 non-amnestic MCI). Plasma biomarkers were measured at baseline and the progression of clinical and cognitive profiles was followed over the three years of follow-up. Baseline plasma NfL concentration increased across the Alzheimer's disease continuum with a mean NfL value of 17.1 ng/mL [SD = 6.1] in non-amnestic MCI, 20.7 ng/mL [SD = 12.0] in amnestic MCI, and 23.1 ng/mL [SD = 22.7] in AD dementia patients. Plasma NfL concentration correlated with age, body mass index (BMI), and global cognitive performance and decline, as measured by the Mini-Mental State Examination (MMSE). MMSE scores decreased in parallel with increasing plasma NfL concentration, independently of age and BMI. However, NfL concentration did not predict MCI participants' conversion to dementia within three years. Discussion: Baseline plasma NfL concentration is associated with cognitive status along the AD continuum, suggesting its usefulness as a potential informative biomarker for cognitive decline follow-up in patients.
Collapse
Affiliation(s)
- Sylvain Lehmann
- Laboratoire et Plateforme de Protéomique Clinique, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, F-34295 Montpellier, France;
| | - Susanna Schraen-Maschke
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, F-59000 Lille, France; (S.S.-M.); (S.B.)
| | - Jean-Sébastien Vidal
- Université Paris Cité, INSERM U1144, GHU APHP Centre, Hopital Broca, Memory Resource and Research Centre de Paris-Broca-Ile de France, F-75013 Paris, France; (J.-S.V.); (O.H.)
| | - Frédéric Blanc
- Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Memory Resource and Research, French National Centre for Scientific Research (CNRS), ICube Laboratory UMR7357 and Fédération de Médecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Intégrative en Santé (IMIS), F-67000 Strasbourg, France;
| | - Claire Paquet
- Université Paris Cité, INSERM U1144, GHU APHP Nord Lariboisière Fernand Widal, Centre de Neurologie Cognitive, F-75010 Paris, France;
| | - Bernadette Allinquant
- Université Paris Cité, Institute of Psychiatry and Neurosciences, Inserm, UMR-S 1266, F-75014 Paris, France;
| | - Stéphanie Bombois
- Univ. Lille, Inserm, CHU Lille, UMR-S-U1172, LiCEND, Lille Neuroscience & Cognition, LabEx DISTALZ, F-59000 Lille, France; (S.S.-M.); (S.B.)
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neurologie, Centre des Maladies Cognitives et Comportementales, GH Pitié-Salpêtrière, F-75013 Paris, France
| | - Audrey Gabelle
- Université de Montpellier, CHU Montpellier, Memory Research and Resources Center, Department of Neurology, Inserm INM NeuroPEPs Team, Excellence Center of Neurodegenerative Disorders, F-34000 Montpellier, France;
| | - Constance Delaby
- Laboratoire et Plateforme de Protéomique Clinique, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, 80 av Fliche, F-34295 Montpellier, France;
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, F-08041 Barcelona, Spain
| | - Olivier Hanon
- Université Paris Cité, INSERM U1144, GHU APHP Centre, Hopital Broca, Memory Resource and Research Centre de Paris-Broca-Ile de France, F-75013 Paris, France; (J.-S.V.); (O.H.)
| |
Collapse
|
25
|
Hazan J, Liu KY, Fox NC, Howard R. Online clinical tools to support the use of new plasma biomarker diagnostic technology in the assessment of Alzheimer's disease: a narrative review. Brain Commun 2023; 5:fcad322. [PMID: 38090277 PMCID: PMC10715781 DOI: 10.1093/braincomms/fcad322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/11/2023] [Accepted: 11/23/2023] [Indexed: 02/15/2024] Open
Abstract
Recent advances in new diagnostic technologies for Alzheimer's disease have improved the speed and precision of diagnosis. However, accessing the potential benefits of this technology poses challenges for clinicians, such as deciding whether it is clinically appropriate to order a diagnostic test, which specific test or tests to order and how to interpret test results and communicate these to the patient and their caregiver. Tools to support decision-making could provide additional structure and information to the clinical assessment process. These tools could be accessed online, and such 'e-tools' can provide an interactive interface to support patients and clinicians in the use of new diagnostic technologies for Alzheimer's disease. We performed a narrative review of the literature to synthesize information available on this research topic. Relevant studies that provide an understanding of how these online tools could be used to optimize the clinical utility of diagnostic technology were identified. Based on these, we discuss the ways in which e-tools have been used to assist in the diagnosis of Alzheimer's disease and propose recommendations for future research to aid further development.
Collapse
Affiliation(s)
- Jemma Hazan
- Division of Psychiatry, University College London, London W1T 7BN, UK
| | - Kathy Y Liu
- Division of Psychiatry, University College London, London W1T 7BN, UK
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, W1T 7NF, UK
| | - Robert Howard
- Division of Psychiatry, University College London, London W1T 7BN, UK
| |
Collapse
|
26
|
Duchateau L, Küҫükali F, De Roeck A, Wittens MMJ, Temmerman J, Weets I, Timmers M, Engelborghs S, Bjerke M, Sleegers K. CSF biomarker analysis of ABCA7 mutation carriers suggests altered APP processing and reduced inflammatory response. Alzheimers Res Ther 2023; 15:195. [PMID: 37946268 PMCID: PMC10634183 DOI: 10.1186/s13195-023-01338-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND The Alzheimer's disease (AD) risk gene ABCA7 has suggested functions in lipid metabolism and the immune system. Rare premature termination codon (PTC) mutations and an expansion of a variable number of tandem repeats (VNTR) polymorphism in the gene, both likely cause a lower ABCA7 expression and hereby increased risk for AD. However, the exact mechanism of action remains unclear. By studying CSF biomarkers reflecting different types of AD-related pathological processes, we aim to get a better insight in those processes and establish a biomarker profile of mutation carriers. METHODS The study population consisted of 229 AD patients for whom CSF was available and ABCA7 sequencing and VNTR genotyping had been performed. This included 28 PTC mutation and 16 pathogenic expansion carriers. CSF levels of Aβ1-42, Aβ1-40, P-tau181, T-tau, sAPPα, sAPPβ, YKL-40, and hFABP were determined using ELISA and Meso Scale Discovery assays. We compared differences in levels of these biomarkers and the Aβ ratio between AD patients with or without an ABCA7 PTC mutation or expansion using linear regression on INT-transformed data with APOE-status, age and sex as covariates. RESULTS Carriers of ABCA7 expansion mutations had significantly lower Aβ1-42 levels (P = 0.022) compared with non-carrier patients. The effect of the presence of ABCA7 mutations on CSF levels was especially pronounced in APOE ε4-negative carriers. In addition, VNTR expansion carriers had reduced Aβ1-40 (P = 0.023), sAPPα (P = 0.047), sAPPβ (P = 0.016), and YKL-40 (P = 0.0036) levels. CONCLUSIONS Our results are suggestive for an effect on APP processing by repeat expansions given the changes in the amyloid-related CSF biomarkers that were found in carriers. The decrease in YKL-40 levels in expansion carriers moreover suggests that these patients potentially have a reduced inflammatory response to AD damage. Moreover, our findings suggest the existence of a mechanism, independent of lowered expression, affecting neuropathology in expansion carriers.
Collapse
Affiliation(s)
- Lena Duchateau
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
| | - Fahri Küҫükali
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
| | - Arne De Roeck
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Present Address: Argenx, Ghent, Belgium
| | - Mandy M J Wittens
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Clinical Neurochemistry Laboratory, Department of Clinical Biology, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
| | - Joke Temmerman
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Jette, Brussels, 1090, Belgium
| | - Ilse Weets
- Clinical Neurochemistry Laboratory, Department of Clinical Biology, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
- Experimental Pharmacology (EFAR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Maarten Timmers
- Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, 2340, Belgium
| | - Sebastiaan Engelborghs
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Jette, Brussels, 1090, Belgium
- Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Department of Neurology and Bru-BRAIN, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
| | - Maria Bjerke
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium
- Clinical Neurochemistry Laboratory, Department of Clinical Biology, University Hospital Brussels, Generaal Jacqueslaan 137, Elsene, Brussels, 1050, Belgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Jette, Brussels, 1090, Belgium
| | - Kristel Sleegers
- Complex Genetics of Alzheimer's Disease Group, VIB-UAntwerp Center for Molecular Neurology, VIB, Building V, Universiteitsplein 1, Wilrijk, Antwerp, B-2610, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp, 2610, Belgium.
| |
Collapse
|
27
|
Satake Y, Kanemoto H, Gotoh S, Akamine S, Suehiro T, Matsunaga K, Shimosegawa E, Yoshiyama K, Morihara T, Mori K, Ikeda M. Cerebrospinal fluid amyloid beta with amyloid positron emission tomography concordance rates in a heterogeneous group of patients including late-onset psychotic disorders: a retrospective cross-sectional study. Psychogeriatrics 2023; 23:1091-1093. [PMID: 37700563 DOI: 10.1111/psyg.13024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Affiliation(s)
- Yuto Satake
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideki Kanemoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shiho Gotoh
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shoshin Akamine
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Suehiro
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Keiko Matsunaga
- Department of Molecular Imaging in Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Eku Shimosegawa
- Department of Molecular Imaging in Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kenji Yoshiyama
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Morihara
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Psychiatry, Toyonaka Municipal Hospital, Toyonaka, Japan
| | - Kohji Mori
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
28
|
Giuffrè GM, Quaranta D, Costantini EM, Citro S, Martellacci N, De Ninno G, Vita MG, Guglielmi V, Rossini PM, Calabresi P, Marra C. Cerebrospinal fluid neurofilament light chain and total-tau as biomarkers of neurodegeneration in Alzheimer's disease and frontotemporal dementia. Neurobiol Dis 2023; 186:106267. [PMID: 37652185 DOI: 10.1016/j.nbd.2023.106267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
INTRODUCTION CSF Neurofilament light chain(NfL) is a promising biomarker of neurodegeneration, but its utility in discriminating between Alzheimer's disease(AD) and frontotemporal dementia(FTD) is limited. METHODS 105 patients with clinical-biological diagnosis of mild cognitive impairment(MCI) due to AD (N = 72) or clinical diagnosis of FTD (N = 33) underwent neuropsychological assessment and CSF Aβ42/40, p-tau181, total-tau and NfL quantification. Group comparisons, correlations between continuous variables and ROC curve analysis were carried out to assess NfL role in discriminating between MCI due to AD and FTD, exploring the associations between NfL, ATN biomarkers and neuropsychological measures. RESULTS NfL levels were significantly lower in the AD group, while levels of total-tau were higher. In the FTD group, significant correlations were found between NfL, p-tau181 and total-tau, and between NfL and cognitive performances. In the AD group, NfL levels were directly correlated with total-tau and p-tau181; Aβ42/40 ratio was inversely correlated with total-tau and p-tau181, but not with NfL. Moreover, p-tau181 and t-tau levels were found to be associated with episodic memory and lexical-semantic impairment. Total-tau/NfL ratio differentiated prodromal-AD from FTD with an AUC of 0.951, higher than the individual measures. DISCUSSION & CONCLUSIONS The results support that NfL and total-tau levels reflect distinct pathophysiological neurodegeneration mechanisms, independent and dependent of Aβ pathology, respectively, Combining them may enhance both markers reliability, their ratio showing high accuracy in distinguishing MCI due to AD from FTD. Moreover, our results revealed associations between NfL and disease severity in FTD and between tauopathy and episodic memory and lexical-semantic impairment in prodromal-AD.
Collapse
Affiliation(s)
- Guido Maria Giuffrè
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Davide Quaranta
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy.
| | | | - Salvatore Citro
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Noemi Martellacci
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Grazia De Ninno
- UOC of Chemistry, Biochemistry and Clinical Molecular Biology - Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Gabriella Vita
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valeria Guglielmi
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Rome, Italy
| | - Paolo Calabresi
- Neurology Unit Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| | - Camillo Marra
- Memory Clinic Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
29
|
Brum WS, Cullen NC, Janelidze S, Ashton NJ, Zimmer ER, Therriault J, Benedet AL, Rahmouni N, Tissot C, Stevenson J, Servaes S, Triana-Baltzer G, Kolb HC, Palmqvist S, Stomrud E, Rosa-Neto P, Blennow K, Hansson O. A two-step workflow based on plasma p-tau217 to screen for amyloid β positivity with further confirmatory testing only in uncertain cases. NATURE AGING 2023; 3:1079-1090. [PMID: 37653254 PMCID: PMC10501903 DOI: 10.1038/s43587-023-00471-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
Cost-effective strategies for identifying amyloid-β (Aβ) positivity in patients with cognitive impairment are urgently needed with recent approvals of anti-Aβ immunotherapies for Alzheimer's disease (AD). Blood biomarkers can accurately detect AD pathology, but it is unclear whether their incorporation into a full diagnostic workflow can reduce the number of confirmatory cerebrospinal fluid (CSF) or positron emission tomography (PET) tests needed while accurately classifying patients. We evaluated a two-step workflow for determining Aβ-PET status in patients with mild cognitive impairment (MCI) from two independent memory clinic-based cohorts (n = 348). A blood-based model including plasma tau protein 217 (p-tau217), age and APOE ε4 status was developed in BioFINDER-1 (area under the curve (AUC) = 89.3%) and validated in BioFINDER-2 (AUC = 94.3%). In step 1, the blood-based model was used to stratify the patients into low, intermediate or high risk of Aβ-PET positivity. In step 2, we assumed referral only of intermediate-risk patients to CSF Aβ42/Aβ40 testing, whereas step 1 alone determined Aβ-status for low- and high-risk groups. Depending on whether lenient, moderate or stringent thresholds were used in step 1, the two-step workflow overall accuracy for detecting Aβ-PET status was 88.2%, 90.5% and 92.0%, respectively, while reducing the number of necessary CSF tests by 85.9%, 72.7% and 61.2%, respectively. In secondary analyses, an adapted version of the BioFINDER-1 model led to successful validation of the two-step workflow with a different plasma p-tau217 immunoassay in patients with cognitive impairment from the TRIAD cohort (n = 84). In conclusion, using a plasma p-tau217-based model for risk stratification of patients with MCI can substantially reduce the need for confirmatory testing while accurately classifying patients, offering a cost-effective strategy to detect AD in memory clinic settings.
Collapse
Affiliation(s)
- Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia, South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biological Sciences: Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- McGill Centre for Studies in Aging, McGill University, Montreal, Québec, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | | | - Hartmuth C Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
30
|
Nisenbaum L, Martone R, Chen T, Rajagovindan R, Dent G, Beaver J, Rubel C, Racine A, He P, Harrison K, Dean R, Vandijck M, Haeberlein SB. CSF biomarker concordance with amyloid PET in Phase 3 studies of aducanumab. Alzheimers Dement 2023; 19:3379-3388. [PMID: 36795603 DOI: 10.1002/alz.12919] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 02/17/2023]
Abstract
INTRODUCTION We assessed the use of cerebrospinal fluid (CSF) biomarkers as an alternative to positron emission tomography (PET) for brain amyloid beta (Aβ) pathology confirmation in the EMERGE and ENGAGE clinical trials. METHODS EMERGE and ENGAGE were randomized, placebo-controlled, Phase 3 trials of aducanumab in participants with early Alzheimer's disease. Concordance between CSF biomarkers (Aβ42, Aβ40, phosphorylated tau 181, and total tau) and amyloid PET status (visual read) at screening was examined. RESULTS Robust concordance between CSF biomarkers and amyloid PET visual status was observed (for Aβ42/Aβ40, AUC: 0.90; 95% CI: 0.83-0.97; p < 0.0001), confirming CSF biomarkers as a reliable alternative to amyloid PET in these studies. Compared with single CSF biomarkers, CSF biomarker ratios showed better agreement with amyloid PET visual reads, demonstrating high diagnostic accuracy. DISCUSSION These analyses add to the growing body of evidence supporting CSF biomarkers as reliable alternatives to amyloid PET imaging for brain Aβ pathology confirmation. HIGHLIGHTS CSF biomarkers and amyloid PET concordance were assessed in Ph3 aducanumab trials. Robust concordance between CSF biomarkers and amyloid PET was observed. CSF biomarker ratios increased diagnostic accuracy over single CSF biomarkers. CSF Aβ42/Aβ40 demonstrated high concordance with amyloid PET. Results support CSF biomarker testing as a reliable alternative to amyloid PET.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ping He
- Biogen, Cambridge, Massachusetts, USA
| | | | - Robert Dean
- Robert A. Dean Consulting, LLC, Indianapolis, Indiana, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
31
|
Wuestefeld A, Pichet Binette A, Berron D, Spotorno N, van Westen D, Stomrud E, Mattsson-Carlgren N, Strandberg O, Smith R, Palmqvist S, Glenn T, Moes S, Honer M, Arfanakis K, Barnes LL, Bennett DA, Schneider JA, Wisse LEM, Hansson O. Age-related and amyloid-beta-independent tau deposition and its downstream effects. Brain 2023; 146:3192-3205. [PMID: 37082959 PMCID: PMC10393402 DOI: 10.1093/brain/awad135] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 04/22/2023] Open
Abstract
Amyloid-β (Aβ) is hypothesized to facilitate the spread of tau pathology beyond the medial temporal lobe. However, there is evidence that, independently of Aβ, age-related tau pathology might be present outside of the medial temporal lobe. We therefore aimed to study age-related Aβ-independent tau deposition outside the medial temporal lobe in two large cohorts and to investigate potential downstream effects of this on cognition and structural measures. We included 545 cognitively unimpaired adults (40-92 years) from the BioFINDER-2 study (in vivo) and 639 (64-108 years) from the Rush Alzheimer's Disease Center cohorts (ex vivo). 18F-RO948- and 18F-flutemetamol-PET standardized uptake value ratios were calculated for regional tau and global/regional Aβ in vivo. Immunohistochemistry was used to estimate Aβ load and tangle density ex vivo. In vivo medial temporal lobe volumes (subiculum, cornu ammonis 1) and cortical thickness (entorhinal cortex, Brodmann area 35) were obtained using Automated Segmentation for Hippocampal Subfields packages. Thickness of early and late neocortical Alzheimer's disease regions was determined using FreeSurfer. Global cognition and episodic memory were estimated to quantify cognitive functioning. In vivo age-related tau deposition was observed in the medial temporal lobe and in frontal and parietal cortical regions, which was statistically significant when adjusting for Aβ. This was also observed in individuals with low Aβ load. Tau deposition was negatively associated with cortical volumes and thickness in temporal and parietal regions independently of Aβ. The associations between age and cortical volume or thickness were partially mediated via tau in regions with early Alzheimer's disease pathology, i.e. early tau and/or Aβ pathology (subiculum/Brodmann area 35/precuneus/posterior cingulate). Finally, the associations between age and cognition were partially mediated via tau in Brodmann area 35, even when including Aβ-PET as covariate. Results were validated in the ex vivo cohort showing age-related and Aβ-independent increases in tau aggregates in and outside the medial temporal lobe. Ex vivo age-cognition associations were mediated by medial and inferior temporal tau tangle density, while correcting for Aβ density. Taken together, our study provides support for primary age-related tauopathy even outside the medial temporal lobe in vivo and ex vivo, with downstream effects on structure and cognition. These results have implications for our understanding of the spreading of tau outside the medial temporal lobe, also in the context of Alzheimer's disease. Moreover, this study suggests the potential utility of tau-targeting treatments in primary age-related tauopathy, likely already in preclinical stages in individuals with low Aβ pathology.
Collapse
Affiliation(s)
- Anika Wuestefeld
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Danielle van Westen
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, SE-222 42 Lund, Sweden
- Image and Function, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Department of Neurology, Skåne University Hospital, SE-205 02 Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Department of Neurology, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Trevor Glenn
- Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Svenja Moes
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Michael Honer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Lisa L Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Laura E M Wisse
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, SE-222 42 Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| |
Collapse
|
32
|
Paciotti S, Wojdała AL, Bellomo G, Toja A, Chipi E, Piersma SR, Pham TV, Gaetani L, Jimenez CR, Parnetti L, Chiasserini D. Potential diagnostic value of CSF metabolism-related proteins across the Alzheimer's disease continuum. Alzheimers Res Ther 2023; 15:124. [PMID: 37454217 DOI: 10.1186/s13195-023-01269-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) cerebrospinal fluid (CSF) core biomarkers (Aβ42/40 ratio, p-tau, and t-tau) provide high diagnostic accuracy, even at the earliest stage of disease. However, these markers do not fully reflect the complex AD pathophysiology. Recent large scale CSF proteomic studies revealed several new AD candidate biomarkers related to metabolic pathways. In this study we measured the CSF levels of four metabolism-related proteins not directly linked to amyloid- and tau-pathways (i.e., pyruvate kinase, PKM; aldolase, ALDO; ubiquitin C-terminal hydrolase L1, UCHL1, and fatty acid-binding protein 3, FABP3) across the AD continuum. We aimed at validating the potential value of these proteins as new CSF biomarkers for AD and their possible involvement in AD pathogenesis, with specific interest on the preclinical phase of the disease. METHODS CSF PKM and ALDO activities were measured with specific enzyme assays while UCHL1 and FABP3 levels were measured with immunoassays in a cohort of patients composed as follows: preclinical AD (pre-AD, n = 19, cognitively unimpaired), mild cognitive impairment due to AD (MCI-AD, n = 50), dementia due to AD (ADdem, n = 45), and patients with frontotemporal dementia (FTD, n = 37). Individuals with MCI not due to AD (MCI, n = 30) and subjective cognitive decline (SCD, n = 52) with negative CSF AD-profile, were enrolled as control groups. RESULTS CSF UCHL1 and FABP3 levels, and PKM activity were significantly increased in AD patients, already at the pre-clinical stage. CSF PKM activity was also increased in FTD patients compared with control groups, being similar between AD and FTD patients. No difference was found in ALDO activity among the groups. UCHL1 showed good performance in discriminating early AD patients (pre-AD and MCI-AD) from controls (AUC ~ 0.83), as assessed by ROC analysis. Similar results were obtained for FABP3. Conversely, PKM provided the best performance when comparing FTD vs. MCI (AUC = 0.80). Combination of PKM, FABP3, and UCHL1 improved the diagnostic accuracy for the detection of patients within the AD continuum when compared with single biomarkers. CONCLUSIONS Our study confirmed the potential role of UCHL1 and FABP3 as neurodegenerative biomarkers for AD. Furthermore, our results validated the increase of PKM activity in CSF of AD patients, already at the preclinical phase of the disease. Increased PKM activity was observed also in FTD patients, possibly underlining similar alterations in energy metabolism in AD and FTD.
Collapse
Affiliation(s)
- Silvia Paciotti
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Anna Lidia Wojdała
- Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Toja
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Chipi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sander R Piersma
- OncoProteomics Laboratory, Laboratory Medical Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Thang V Pham
- OncoProteomics Laboratory, Laboratory Medical Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Connie R Jimenez
- OncoProteomics Laboratory, Laboratory Medical Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
33
|
Kodosaki E, Zetterberg H, Heslegrave A. Validating blood tests as a possible routine diagnostic assay of Alzheimer's disease. Expert Rev Mol Diagn 2023; 23:1153-1165. [PMID: 38018372 DOI: 10.1080/14737159.2023.2289553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION In recent years, exciting developments in disease modifying treatments for Alzheimer's disease (AD) have made accurate and timely diagnosis of this disease a priority. Blood biomarkers (BBMs) for amyloid pathology using improved immunoassay and mass spectrometry techniques have been an area of intense research for the last 10 years and are coming to the fore, as a real prospect to be used in the clinical diagnostics of the disease. AREAS COVERED The following review will update and discuss blood biomarkers that will be most useful in diagnosing AD and the context necessary for their implementation. EXPERT OPINION It is clear we now have BBMs, and technology to measure them, that are capable of detecting amyloid pathology in AD. The challenge is to validate them across platforms and populations to incorporate them into clinical practice. It is important that implementation comes with education, we need to give clinicians the tools for appropriate use and interpretation. It is feasible that BBMs will be used to screen populations, initially for clinical trial entry but also therapeutic intervention in the foreseeable future. We now need to focus BBM research on other pathologies to ensure we accelerate the development of therapeutics for all neurodegenerative diseases.
Collapse
Affiliation(s)
- Eleftheria Kodosaki
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Dementia Research Institute at UCL, London, UK
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Dementia Research Institute at UCL, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wisconsin Alzheimer's Disease Research Centre, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology,Dementia Research Institute at UCL, London, UK
- Hong Kong Centre for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Dementia Research Institute at UCL, London, UK
| |
Collapse
|
34
|
Gaetani L, Chiasserini D, Paolini Paoletti F, Bellomo G, Parnetti L. Required improvements for cerebrospinal fluid-based biomarker tests of Alzheimer's disease. Expert Rev Mol Diagn 2023; 23:1195-1207. [PMID: 37902844 DOI: 10.1080/14737159.2023.2276918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/25/2023] [Indexed: 11/01/2023]
Abstract
INTRODUCTION Cerebrospinal fluid (CSF) biomarkers represent a well-established tool for diagnosing Alzheimer's disease (AD), independently from the clinical stage, by reflecting the presence of brain amyloidosis (A+) and tauopathy (T+). In front of this important achievement, so far, (i) CSF AD biomarkers have not yet been adopted for routine clinical use in all Centers dedicated to AD, mainly due to inter-lab variation and lack of internationally accepted cutoff values; (ii) we do need to add other biomarkers more suitable to correlate with the clinical stage and disease monitoring; (iii) we also need to detect the co-presence of other 'non-AD' pathologies. AREAS COVERED Efforts to establish standardized cutoff values based on large-scale multi-center studies are discussed. The influence of aging and comorbidities on CSF biomarker levels is also analyzed, and possible solutions are presented, i.e. complementing the A/T/(N) system with markers of axonal damage and synaptic derangement. EXPERT OPINION The first, mandatory need is to reach common cutoff values and defined (automated) methodologies for CSF AD biomarkers. To properly select subjects deserving CSF analysis, blood tests might represent the first-line approach. In those subjects undergoing CSF analysis, multiple biomarkers, able to give a comprehensive and personalized pathophysiological/prognostic information, should be included.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
35
|
Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer's disease in clinical practice and trials. NATURE AGING 2023; 3:506-519. [PMID: 37202517 PMCID: PMC10979350 DOI: 10.1038/s43587-023-00403-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Blood-based biomarkers hold great promise to revolutionize the diagnostic and prognostic work-up of Alzheimer's disease (AD) in clinical practice. This is very timely, considering the recent development of anti-amyloid-β (Aβ) immunotherapies. Several assays for measuring phosphorylated tau (p-tau) in plasma exhibit high diagnostic accuracy in distinguishing AD from all other neurodegenerative diseases in patients with cognitive impairment. Prognostic models based on plasma p-tau levels can also predict future development of AD dementia in patients with mild cognitive complaints. The use of such high-performing plasma p-tau assays in the clinical practice of specialist memory clinics would reduce the need for more costly investigations involving cerebrospinal fluid samples or positron emission tomography. Indeed, blood-based biomarkers already facilitate identification of individuals with pre-symptomatic AD in the context of clinical trials. Longitudinal measurements of such biomarkers will also improve the detection of relevant disease-modifying effects of new drugs or lifestyle interventions.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Lund, 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
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for 27 Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeffrey Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
36
|
Musso G, Cosma C, Zaninotto M, Gabelli C, Basso D, Plebani M. Pre-analytical variability of the Lumipulse immunoassay for plasma biomarkers of Alzheimer's disease. Clin Chem Lab Med 2023; 61:e53-e56. [PMID: 36423341 DOI: 10.1515/cclm-2022-0770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Giulia Musso
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy.,Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy.,QI.LAB.MED, Spin-off of the University of Padova, Padova, Italy
| | - Martina Zaninotto
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy.,QI.LAB.MED, Spin-off of the University of Padova, Padova, Italy
| | - Carlo Gabelli
- Regional Brain Aging Center, University-Hospital of Padova, Padova, Italy
| | - Daniela Basso
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy.,Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Mario Plebani
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy.,Department of Medicine-DIMED, University of Padova, Padova, Italy.,QI.LAB.MED, Spin-off of the University of Padova, Padova, Italy
| |
Collapse
|
37
|
Zou Y, Yu S, Ma X, Ma C, Mao C, Mu D, Li L, Gao J, Qiu L. How far is the goal of applying β-amyloid in cerebrospinal fluid for clinical diagnosis of Alzheimer's disease with standardization of measurements? Clin Biochem 2023; 112:33-42. [PMID: 36473516 DOI: 10.1016/j.clinbiochem.2022.11.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/02/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Cerebrospinal fluid (CSF) β-amyloid (Aβ) is important for early diagnosis of Alzheimer's disease (AD). However, the cohort distributions and cut-off values have large variation across different analytical assays, kits, and laboratories. In this review, we summarize the cut-off values and diagnostic performance for CSF Aβ1-42 and Aβ1-42/Aβ1-40, and explore the important effect factors. Based on the Alzheimer's Association external quality control program (AAQC program), the peer group coefficient of variation of manual ELISA assays for CSF Aβ1-42 was unsatisfied (>20%). Fully automated platforms with better performance have recently been developed, but still not widely applied. In 2020, the certified reference material (CRM) for CSF Aβ1-42 was launched; however, the AAQC 2021-round results did not show effective improvements. Thus, further development and popularization of CRM for CSF Aβ1-42 and Aβ1-40 are urgently required. Standardizing the diagnostic procedures of AD and related status and the pre-analytical protocols of CSF samples, improving detection performance of analytical assays, and popularizing the application of fully automated platforms are also important for the establishment of uniform cut-off values. Moreover, each laboratory should verify the applicability of uniform cut-off values, and evaluate whether it is necessary to establish its own population- and assay-specific cut-off values.
Collapse
Affiliation(s)
- Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Songlin Yu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Xiaoli Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China; Medical Science Research Center, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Chaochao Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Chenhui Mao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Danni Mu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Lei Li
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Jing Gao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
38
|
Johnson SC, Suárez-Calvet M, Suridjan I, Minguillón C, Gispert JD, Jonaitis E, Michna A, Carboni M, Bittner T, Rabe C, Kollmorgen G, Zetterberg H, Blennow K. Identifying clinically useful biomarkers in neurodegenerative disease through a collaborative approach: the NeuroToolKit. Alzheimers Res Ther 2023; 15:25. [PMID: 36709293 PMCID: PMC9883877 DOI: 10.1186/s13195-023-01168-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 01/15/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex and heterogeneous disease, which requires reliable biomarkers for diagnosis and monitoring disease activity. Preanalytical protocol and technical variability associated with biomarker immunoassays makes comparability of biomarker data across multiple cohorts difficult. This study aimed to compare cerebrospinal fluid (CSF) biomarker results across independent cohorts, including participants spanning the AD continuum. METHODS Measured on the NeuroToolKit (NTK) prototype panel of immunoassays, 12 CSF biomarkers were evaluated from three cohorts (ALFA+, Wisconsin, and Abby/Blaze). A correction factor was applied to biomarkers found to be affected by preanalytical procedures (amyloid-β1-42, amyloid-β1-40, and alpha-synuclein), and results between cohorts for each disease stage were compared. The relationship between CSF biomarker concentration and cognitive scores was evaluated. RESULTS Biomarker distributions were comparable across cohorts following correction. Correlations of biomarker values were consistent across cohorts, regardless of disease stage. Disease stage differentiation was highest for neurofilament light (NfL), phosphorylated tau, and total tau, regardless of the cohort. Correlation between biomarker concentration and cognitive scores was comparable across cohorts, and strongest for NfL, chitinase-3-like protein-1 (YKL40), and glial fibrillary acidic protein. DISCUSSION The precision of the NTK enables merging of biomarker datasets, after correction for preanalytical confounders. Assessment of multiple cohorts is crucial to increase power in future studies into AD pathogenesis.
Collapse
Affiliation(s)
- Sterling C Johnson
- University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center of the William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005, Barcelona, Spain.
- Centre for Biomedical Research Network on Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
- Neurology Service, Hospital del Mar, Barcelona, Spain.
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.
| | | | - Carolina Minguillón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005, Barcelona, Spain
- Centre for Biomedical Research Network on Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centre for Biomedical Research in Network Bioengineering, Biomaterials and Nanomedicine, Instituto de Salud Carlos III, Madrid, Spain
| | - Erin Jonaitis
- University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Tobias Bittner
- F. Hoffmann-La Roche AG, Basel, Switzerland
- Genentech, A Member of the Roche Group, San Francisco, CA, USA
| | - Christina Rabe
- Genentech, A Member of the Roche Group, San Francisco, CA, USA
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
39
|
Blennow K, Stomrud E, Zetterberg H, Borlinghaus N, Corradini V, Manuilova E, Müller-Hübner L, Quevenco FC, Rutz S, Hansson O. Second-generation Elecsys cerebrospinal fluid immunoassays aid diagnosis of early Alzheimer's disease. Clin Chem Lab Med 2023; 61:234-244. [PMID: 36282960 DOI: 10.1515/cclm-2022-0516] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/10/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Timely diagnosis of Alzheimer's disease (AD) is critical for appropriate treatment/patient management. Cerebrospinal fluid (CSF) biomarker analysis is often used to aid diagnosis. We assessed analytical performance of second-generation (Gen II) Elecsys® CSF immunoassays (Roche Diagnostics International Ltd), and adjusted existing cut-offs, to evaluate their potential utility in clinical routine. METHODS Analytical performance was assessed using CSF samples measured with Elecsys CSF Gen II immunoassays on cobas e analyzers. Aβ42 Gen I/Gen II immunoassay method comparisons were performed (Passing-Bablok regression). Cut-off values were adjusted using estimated bias in biomarker levels between BioFINDER protocol aliquots/Gen I immunoassays and Gen II protocol aliquots/immunoassays. Distribution of Gen II immunoassay values was evaluated in AD, mild cognitive impairment (MCI), and cognitively normal cohorts; percentage observations outside the measuring range were derived. RESULTS The Gen II immunoassays demonstrated good analytical performance, including repeatability, intermediate precision, lot-to-lot agreement (Pearson's r: ≥0.999), and platform agreement (Pearson's r: ≥0.995). Aβ42 Gen I/Gen II immunoassay measurements were strongly correlated (Pearson's r: 0.985-0.999). Aβ42 Gen II immunoassay cut-offs were adjusted to 1,030 and 800 ng/L, and pTau181/Aβ42 ratio cut-offs to 0.023 and 0.029, for Gen II and I protocols, respectively. No observations were below the lower limit of the measuring range; above the upper limit, there were none from the AD cohort, and 2.6 and 6.8% from the MCI and cognitively normal cohorts, respectively. CONCLUSIONS Our findings suggest that the Gen II immunoassays have potential utility in clinical routine to aid diagnosis of AD.
Collapse
Affiliation(s)
- 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
| | - Erik Stomrud
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Malmö, 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
- Hong Kong Center for Neurodegenerative Disease, Shatin, N.T., Hong Kong, P.R. China
| | | | | | | | | | | | | | - Oskar Hansson
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
40
|
Leuzy A, Mattsson-Carlgren N, Cullen NC, Stomrud E, Palmqvist S, La Joie R, Iaccarino L, Zetterberg H, Rabinovici G, Blennow K, Janelidze S, Hansson O. Robustness of CSF Aβ42/40 and Aβ42/P-tau181 measured using fully automated immunoassays to detect AD-related outcomes. Alzheimers Dement 2023. [PMID: 36681387 DOI: 10.1002/alz.12897] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 01/23/2023]
Abstract
INTRODUCTION This study investigated the comparability of cerebrospinal fluid (CSF) cutoffs for Elecsys immunoassays for amyloid beta (Aβ)42/Aβ40 or Aβ42/phosphorylated tau (p-tau)181 and the effects of measurement variability when predicting Alzheimer's disease (AD)-related outcomes (i.e., Aβ-positron emission tomography [PET] visual read and AD neuropathology). METHODS We studied 750 participants (BioFINDER study, Alzheimer's Disease Neuroimaging Initiative [ADNI], and University of California San Francisco [UCSF]). Youden's index was used to identify cutoffs and to calculate accuracy (Aβ-PET visual read as outcome). Using longitudinal variability in Aβ-negative controls, we identified a gray zone around cut-points where the risk of an inconsistent predicted outcome was >5%. RESULTS For Aβ42/Aβ40, cutoffs across cohorts were <0.059 (BioFINDER), <0.057 (ADNI), and <0.058 (UCSF). For Aβ42/p-tau181, cutoffs were <41.90 (BioFINDER), <39.20 (ADNI), and <46.02 (UCSF). Accuracy was ≈90% for both Aβ42/Aβ40 and Aβ42/p-tau181 using these cutoffs. Using Aβ-PET as an outcome, 8.7% of participants fell within a gray zone interval for Aβ42/Aβ40, compared to 4.5% for Aβ42/p-tau181. Similar findings were observed using a measure of overall AD neuropathologic change (7.7% vs. 3.3%). In a subset with CSF and plasma Aβ42/40, the number of individuals within the gray zone was ≈1.5 to 3 times greater when using plasma Aβ42/40. DISCUSSION CSF Aβ42/p-tau181 was more robust to the effects of measurement variability, suggesting that it may be the preferred Elecsys-based measure in clinical practice and trials.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - 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 UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Gil Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - 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
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
41
|
Kurz C, Stöckl L, Schrurs I, Suridjan I, Gürsel SÜ, Bittner T, Jethwa A, Perneczky R. Impact of pre-analytical sample handling factors on plasma biomarkers of Alzheimer's disease. J Neurochem 2023; 165:95-105. [PMID: 36625424 DOI: 10.1111/jnc.15757] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/07/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023]
Abstract
An unmet need exists for reliable plasma biomarkers of amyloid pathology, in the clinical laboratory setting, to streamline diagnosis of Alzheimer's disease (AD). For routine clinical use, a biomarker must provide robust and reliable results under pre-analytical sample handling conditions. We investigated the impact of different pre-analytical sample handling procedures on the levels of seven plasma biomarkers in development for potential routine use in AD. Using (1) fresh (never frozen) and (2) previously frozen plasma, we evaluated the effects of (A) storage time and temperature, (B) freeze/thaw (F/T) cycles, (C) anticoagulants, (D) tube transfer, and (E) plastic tube types. Blood samples were prospectively collected from patients with cognitive impairment undergoing investigation in a memory clinic. β-amyloid 1-40 (Aβ40), β-amyloid 1-42 (Aβ42), apolipoprotein E4, glial fibrillary acidic protein, neurofilament light chain, phosphorylated-tau (phospho-tau) 181, and phospho-tau-217 were measured using Elecsys® plasma prototype immunoassays. Recovery signals for each plasma biomarker and sample handling parameter were calculated. For all plasma biomarkers measured, pre-analytical effects were comparable between fresh (never frozen) and previously frozen samples. All plasma biomarkers tested were stable for ≤24 h at 4°C when stored as whole blood and ethylenediaminetetraacetic acid (EDTA) plasma. Recovery signals were acceptable for up to five tube transfers, or two F/T cycles, and in both polypropylene and low-density polyethylene tubes. For all plasma biomarkers except Aβ42 and Aβ40, analyte levels were largely comparable between EDTA, lithium heparin, and sodium citrate tubes. Aβ42 and Aβ40 were most sensitive to pre-analytical handling, and the effects could only be partially compensated by the Aβ42/Aβ40 ratio. We provide recommendations for an optimal sample handling protocol for analysis of plasma biomarkers for amyloid pathology AD, to improve the reproducibility of future studies on plasma biomarkers assays and for potential use in routine clinical practice.
Collapse
Affiliation(s)
- Carolin Kurz
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | | | | | | | - Selim Üstün Gürsel
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,German Center for Neurodegenerative Disorders (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK.,Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| |
Collapse
|
42
|
Hansson O, Edelmayer RM, Boxer AL, Carrillo MC, Mielke MM, Rabinovici GD, Salloway S, Sperling R, Zetterberg H, Teunissen CE. The Alzheimer's Association appropriate use recommendations for blood biomarkers in Alzheimer's disease. Alzheimers Dement 2022; 18:2669-2686. [PMID: 35908251 PMCID: PMC10087669 DOI: 10.1002/alz.12756] [Citation(s) in RCA: 228] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 01/31/2023]
Abstract
Blood-based markers (BBMs) have recently shown promise to revolutionize the diagnostic and prognostic work-up of Alzheimer's disease (AD), as well as to improve the design of interventional trials. Here we discuss in detail further research needed to be performed before widespread use of BBMs. We already now recommend use of BBMs as (pre-)screeners to identify individuals likely to have AD pathological changes for inclusion in trials evaluating disease-modifying therapies, provided the AD status is confirmed with positron emission tomography (PET) or cerebrospinal fluid (CSF) testing. We also encourage studying longitudinal BBM changes in ongoing as well as future interventional trials. However, BBMs should not yet be used as primary endpoints in pivotal trials. Further, we recommend to cautiously start using BBMs in specialized memory clinics as part of the diagnostic work-up of patients with cognitive symptoms and the results should be confirmed whenever possible with CSF or PET. Additional data are needed before use of BBMs as stand-alone diagnostic AD markers, or before considering use in primary care.
Collapse
Affiliation(s)
- Oskar Hansson
- ClinicalMemory Research UnitDepartment of Clinical Sciences MalmöLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | | | - Adam L. Boxer
- Department of NeurologyUniversity of California San FranciscoMemory and Aging CenterSan FranciscoCaliforniaUSA
| | | | - Michelle M. Mielke
- Department of Epidemiology and PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Gil D. Rabinovici
- Department of NeurologyUniversity of California San FranciscoMemory and Aging CenterSan FranciscoCaliforniaUSA
| | - Stephen Salloway
- Departments of Neurology and PsychiatryAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Reisa Sperling
- Centerfor Alzheimer Research and TreatmentBrigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongPeople's Republic of China
| | - Charlotte E. Teunissen
- NeurochemistryLaboratoryDepartment of Clinical ChemistryAmsterdam University Medical CentersVrije UniversiteitAmsterdam NeuroscienceAmsterdamthe Netherlands
| |
Collapse
|
43
|
Abstract
Alzheimer's disease (AD) characterization has progressed from being indexed using clinical symptomatology followed by neuropathological examination at autopsy to in vivo signatures using cerebrospinal fluid (CSF) biomarkers and positron emission tomography. The core AD biomarkers reflect amyloid-β plaques (A), tau pathology (T) and neurodegeneration (N), following the ATN schedule, and are now being introduced into clinical routine practice. This is an important development, as disease-modifying treatments are now emerging. Further, there are now reproducible data on CSF biomarkers which reflect synaptic pathology, neuroinflammation and common co-pathologies. In addition, the development of ultrasensitive techniques has enabled the core CSF biomarkers of AD pathophysiology to be translated to blood (e.g., phosphorylated tau, amyloid-β and neurofilament light). In this chapter, we review where we stand with both core and novel CSF biomarkers, as well as the explosion of data on blood biomarkers. Also, we discuss potential applications in research aiming to better understand the disease, as well as possible use in routine clinical practice and therapeutic trials.
Collapse
Affiliation(s)
- Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Anders Elmgren
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, United Kingdom; UK Dementia Research Institute, University College London, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
44
|
Nojima H, Ito S, Kushida A, Abe A, Motsuchi W, Verbel D, Vandijck M, Jannes G, Vandenbroucke I, Aoyagi K. Clinical utility of cerebrospinal fluid biomarkers measured by LUMIPULSE ® system. Ann Clin Transl Neurol 2022; 9:1898-1909. [PMID: 36321325 PMCID: PMC9735374 DOI: 10.1002/acn3.51681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) are well-established in research settings, but their use in routine clinical practice remains a largely unexploited potential. Here, we examined the relationship between CSF biomarkers, measured by a fully automated immunoassay platform, and brain β-amyloid (Aβ) deposition status confirmed by amyloid positron emission tomography (PET). METHODS One hundred ninety-nine CSF samples from clinically diagnosed AD patients enrolled in a clinical study and who underwent amyloid PET were used for the measurement of CSF biomarkers Aβ 1-40 (Aβ40), Aβ 1-42 (Aβ42), total tau (t-Tau), and phosphorylated tau-181 (p-Tau181) using the LUMIPULSE system. These biomarkers and their combinations were compared to amyloid PET classification (negative or positive) using visual read assessments. Several combinations were also analyzed with a multivariable logistic regression model. RESULTS Aβ42, t-Tau, and p-Tau181, and the ratios of Aβ42 with other biomarkers had a good diagnostic agreement with amyloid PET imaging. The multivariable logistic regression analysis showed that amyloid PET status was associated with Aβ40 and Aβ42, but other factors, such as MMSE, sex, t-Tau, and p-Tau181, did not significantly add information to the model. CONCLUSIONS CSF biomarkers measured with the LUMIPULSE system showed good agreement with amyloid PET imaging. The ratio of Aβ42 with the other analyzed biomarkers showed a higher correlation with amyloid PET than Aβ42 alone, suggesting that the combinations of biomarkers could be useful in the diagnostic assessment in clinical research and potentially in routine clinical practice.
Collapse
Affiliation(s)
- Hisashi Nojima
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - Satoshi Ito
- Eisai Co., Ltd. 4‐6‐10 KoishikawaBunkyo‐kuTokyo112‐8088Japan,Eisai Inc.200 Metro BoulevardNutleyNew Jersey07110USA
| | - Akira Kushida
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - Aki Abe
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - Wataru Motsuchi
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| | - David Verbel
- Eisai Inc.200 Metro BoulevardNutleyNew Jersey07110USA
| | - Manu Vandijck
- Fujirebio‐Europe N.V.Technologiepark 69052GhentBelgium
| | - Geert Jannes
- Fujirebio‐Europe N.V.Technologiepark 69052GhentBelgium
| | | | - Katsumi Aoyagi
- FUJIREBIO Inc.2‐1‐1, Nishishinjuku, Shinjuku‐kuTokyo163‐0410Japan
| |
Collapse
|
45
|
Delaby C, Teunissen CE, Blennow K, Alcolea D, Arisi I, Amar EB, Beaume A, Bedel A, Bellomo G, Bigot‐Corbel E, Bjerke M, Blanc‐Quintin M, Boada M, Bousiges O, Chapman MD, DeMarco ML, D'Onofrio M, Dumurgier J, Dufour‐Rainfray D, Engelborghs S, Esselmann H, Fogli A, Gabelle A, Galloni E, Gondolf C, Grandhomme F, Grau‐Rivera O, Hart M, Ikeuchi T, Jeromin A, Kasuga K, Keshavan A, Khalil M, Körtvelyessy P, Kulczynska‐Przybik A, Laplanche J, Lewczuk P, Li Q, Lleó A, Malaplate C, Marquié M, Masters CL, Mroczko B, Nogueira L, Orellana A, Otto M, Oudart J, Paquet C, Paoletti FP, Parnetti L, Perret‐Liaudet A, Peoc'h K, Poesen K, Puig‐Pijoan A, Quadrio I, Quillard‐Muraine M, Rucheton B, Schraen S, Schott JM, Shaw LM, Suárez‐Calvet M, Tsolaki M, Tumani H, Udeh‐Momoh CT, Vaudran L, Verbeek MM, Verde F, Vermunt L, Vogelgsang J, Wiltfang J, Zetterberg H, Lehmann S. Clinical reporting following the quantification of cerebrospinal fluid biomarkers in Alzheimer's disease: An international overview. Alzheimers Dement 2022; 18:1868-1879. [PMID: 34936194 PMCID: PMC9787404 DOI: 10.1002/alz.12545] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/11/2021] [Accepted: 10/25/2021] [Indexed: 01/28/2023]
Abstract
INTRODUCTION The current practice of quantifying cerebrospinal fluid (CSF) biomarkers as an aid in the diagnosis of Alzheimer's disease (AD) varies from center to center. For a same biochemical profile, interpretation and reporting of results may differ, which can lead to misunderstandings and raises questions about the commutability of tests. METHODS We obtained a description of (pre-)analytical protocols and sample reports from 40 centers worldwide. A consensus approach allowed us to propose harmonized comments corresponding to the different CSF biomarker profiles observed in patients. RESULTS The (pre-)analytical procedures were similar between centers. There was considerable heterogeneity in cutoff definitions and report comments. We therefore identified and selected by consensus the most accurate and informative comments regarding the interpretation of CSF biomarkers in the context of AD diagnosis. DISCUSSION This is the first time that harmonized reports are proposed across worldwide specialized laboratories involved in the biochemical diagnosis of AD.
Collapse
Affiliation(s)
- Constance Delaby
- LBPC‐PPCUniv MontpellierCHU MontpellierINSERMMontpellierFrance,Hospital de la Santa Creu i Sant Pau ‐ Biomedical Research Institute Sant Pau ‐ Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Charlotte E. Teunissen
- Neurochemistry LabDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamNetherlands
| | - Kaj Blennow
- Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Daniel Alcolea
- Hospital de la Santa Creu i Sant Pau ‐ Biomedical Research Institute Sant Pau ‐ Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Ivan Arisi
- European Brain Research Institute (EBRI) “Rita Levi‐Montalcini”RomaItaly
| | - Elodie Bouaziz Amar
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | | | | | - Giovanni Bellomo
- Lab of Clinical NeurochemistrySection of NeurologyDept. of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | | - Maria Bjerke
- Vrije Universiteit BrusselCenter for Neurosciences and Department of Clinical BiologyClinical Neurochemistry LaboratoryUniversitair Ziekenhuis BrusselBrusselsBelgium,Department of Biomedical Sciences, Institute Born‐BungeUniversity of AntwerpAntwerpBelgium
| | | | - Mercè Boada
- Research Center and Memory ClinicFundació ACEInstitut Català de Neurociències Aplicades and Universitat Internacional de Catalunya (UIC)BarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Olivier Bousiges
- Laboratoire de Biochimie et Biologie Moléculaire, et CNRSICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg)Team IMISHôpitaux Universitaires de StrasbourgStrasbourgFrance
| | - Miles D Chapman
- Department of NeuroimmunologyNational Hospital for Neurology and Neurosurgery, UCL Queen SquareLondonUK
| | - Mari L. DeMarco
- Department of Pathology and Laboratory MedicineSt. Paul's Hospital, Providence Health Care, Vancouver, Canada & Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverCanada
| | - Mara D'Onofrio
- European Brain Research Institute (EBRI) “Rita Levi‐Montalcini”RomaItaly
| | - Julien Dumurgier
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | | | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, Institute Born‐BungeUniversity of AntwerpAntwerpBelgium,Vrije Universiteit BrusselUniversitair Ziekenhuis BrusselCenter for Neurosciences and Department of NeurologyBrusselsBelgium
| | - Hermann Esselmann
- Department of Psychiatry and PsychotherapyUniversity Medical Center Goettingen (UMGGoettingenGermany
| | - Anne Fogli
- CHU Clermont‐FerrandClermont‐FerrandFrance
| | - Audrey Gabelle
- LBPC‐PPCUniv MontpellierCHU MontpellierINSERMMontpellierFrance
| | | | | | | | - Oriol Grau‐Rivera
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain,Servei de NeurologiaHospital del MarUnitat de deteriorament cognitiu i transtorns del movimentBarcelonaSpain,IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Melanie Hart
- Department of NeuroimmunologyNational Hospital for Neurology and Neurosurgery, UCL Queen SquareLondonUK
| | - Takeshi Ikeuchi
- Dept. of Molecular GeneticsCenter for BioresourcesBrain Research InstituteNiigata UniversityNiigataJapan
| | | | - Kensaku Kasuga
- Dept. of Molecular GeneticsCenter for BioresourcesBrain Research InstituteNiigata UniversityNiigataJapan
| | - Ashvini Keshavan
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | | | - Peter Körtvelyessy
- Freie Universität Berlin and Humboldt‐Universität zu BerlinDepartment of NeurologyGerman Center for Neurodegenerative Diseases, Magdeburg, Germany and Charité‐Universitäts medizin BerlinBerlinGermany
| | | | - Jean‐Louis Laplanche
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | - Piotr Lewczuk
- Department of Neurodegeneration DiagnosticsMedical University of BialystokBialystokPoland,Lab for Clinical Neurochemistry and Neurochemical Dementia DiagnosticsUniversitätsklinikum Erlangen and Friedrich‐Alexander Universität Erlangen‐NürnbergErlangenGermany
| | - Qiao‐Xin Li
- Florey Institute and The University of MelbourneMelbourneVictoriaAustralia
| | - Alberto Lleó
- Hospital de la Santa Creu i Sant Pau ‐ Biomedical Research Institute Sant Pau ‐ Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Catherine Malaplate
- CHRU de NancyLaboratoire de BiochimieBiologie Moléculaire et Nutrition/ Université de LorraineNancyFrance
| | - Marta Marquié
- Research Center and Memory ClinicFundació ACEInstitut Català de Neurociències Aplicades and Universitat Internacional de Catalunya (UIC)BarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Colin L. Masters
- Florey Institute and The University of MelbourneMelbourneVictoriaAustralia
| | - Barbara Mroczko
- Department of Neurodegeneration DiagnosticsMedical University of BialystokBialystokPoland
| | - Léonor Nogueira
- Laboratoire de Biologie Cellulaire et CytologieCHU PURPANToulouseFrance
| | - Adelina Orellana
- Research Center and Memory ClinicFundació ACEInstitut Català de Neurociències Aplicades and Universitat Internacional de Catalunya (UIC)BarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Markus Otto
- Department of Neurology and CSF LaboratoryUniversity of UlmUlmGermany
| | | | - Claire Paquet
- Université de ParisCognitive Neurology CenterGHU APHP Nord Lariboisière Fernand‐Widal HospitalParisFrance
| | - Federico Paolini Paoletti
- Lab of Clinical NeurochemistrySection of NeurologyDept. of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Lucilla Parnetti
- Lab of Clinical NeurochemistrySection of NeurologyDept. of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Armand Perret‐Liaudet
- Lyon Neuroscience Research Center BIORAN Team ‐ CNRS UMR 5292INSERM U1028Lyon University HospitalLyonFrance
| | - Katell Peoc'h
- Université de Paris GHU APHP Nord Beaujon HospitalParisFrance
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker Research (LaMoN)Department of NeurosciencesKU LeuvenLeuven Brain InstituteLeuvenBelgium
| | - Albert Puig‐Pijoan
- Servei de NeurologiaHospital del MarUnitat de deteriorament cognitiu i transtorns del movimentBarcelonaSpain,IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | - Isabelle Quadrio
- Lyon Neuroscience Research Center BIORAN Team ‐ CNRS UMR 5292INSERM U1028Lyon University HospitalLyonFrance
| | - Muriel Quillard‐Muraine
- UNIROUENRouen University HospitalDepartment of Clinical biologyBiochemistry laboratoryNormandie UnivRouenFrance
| | | | - Susanna Schraen
- InsermCHU LilleU1172‐LilNCogLICENDLabEx DISTALZUniversité de LilleLilleFrance
| | | | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine HospitalUniversity of PennsylvaniaPennsylvaniaUSA
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain,Servei de NeurologiaHospital del MarUnitat de deteriorament cognitiu i transtorns del movimentBarcelonaSpain,IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Magda Tsolaki
- 1st Department of NeurologySchool of MedicineFaculty of Health of SciencesAristotle University of ThessalonikiThessalonikiGreece
| | - Hayrettin Tumani
- Department of Neurology and CSF LaboratoryUniversity of UlmUlmGermany
| | | | | | - Marcel M Verbeek
- Donders Institute for Brain, Cognition and BehaviourRadboud Alzheimer CentreDepartments of Neurology and Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Federico Verde
- Department of Neurology ‐ Stroke Unit and Laboratory of NeuroscienceIRCCS Istituto Auxologico ItalianoMilanItaly,Department of Pathophysiology and Transplantation“Dino Ferrari” Center, Università degli Studi di MilanoMilanItaly
| | - Lisa Vermunt
- Neurochemistry LabDepartment of Clinical ChemistryAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamNetherlands
| | - Jonathan Vogelgsang
- Department of Psychiatry and PsychotherapyUniversity Medical Center Goettingen (UMGGoettingenGermany,McLean HospitalTranslational Neuroscience LaboratoryHarvard Medical SchoolBelmontMassachusettsUSA
| | - Jens Wiltfang
- Department of Psychiatry and PsychotherapyUniversity Medical Center Goettingen (UMGGoettingenGermany,German Center for Neurodegenerative Diseases (DZNE)GoettingenGermany,Neurosciences and Signaling GroupInstitute of Biomedicine (iBiMED)Department of Medical SciencesUniversity of AveiroAveiroPortugal
| | - Henrik Zetterberg
- Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden,UK Dementia Research Institute at UCLLondonUK,Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Sylvain Lehmann
- LBPC‐PPCUniv MontpellierCHU MontpellierINSERMMontpellierFrance
| |
Collapse
|
46
|
Xu C, Zhao L, Dong C. A Review of Application of Aβ42/40 Ratio in Diagnosis and Prognosis of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:495-512. [DOI: 10.3233/jad-220673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of patients with Alzheimer’s disease (AD) and non-Alzheimer’s disease (non-AD) has drastically increased over recent decades. The amyloid cascade hypothesis attributes a vital role to amyloid-β protein (Aβ) in the pathogenesis of AD. As the main pathological hallmark of AD, amyloid plaques consist of merely the 42 and 40 amino acid variants of Aβ (Aβ 42 and Aβ 40). The cerebrospinal fluid (CSF) biomarker Aβ 42/40 has been extensively investigated and eventually integrated into important diagnostic tools to support the clinical diagnosis of AD. With the development of highly sensitive assays and technologies, blood-based Aβ 42/40, which was obtained using a minimally invasive and cost-effective method, has been proven to be abnormal in synchrony with CSF biomarker values. This paper presents the recent progress of the CSF Aβ 42/40 ratio and plasma Aβ 42/40 for AD as well as their potential clinical application as diagnostic markers or screening tools for dementia.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
47
|
Huang S, Wang J, Fan DY, Luo T, Li Y, Tu YF, Shen YY, Zeng GH, Chen DW, Wang YR, Chen LY, Wang YJ, Guo J. The association of serum uric acid with cognitive impairment and ATN biomarkers. Front Aging Neurosci 2022; 14:943380. [PMID: 35923549 PMCID: PMC9339963 DOI: 10.3389/fnagi.2022.943380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background Cognitive impairment (CI) has become a worldwide health problem. The relationship between CI and uric acid (UA) is contradictory. Objective We included participants with a full spectrum of CI, from cognitively unimpaired (CU) to dementia, from the Chongqing Ageing & Dementia Study (CADS). Methods First, we identified the relationships between serum UA (sUA) and cognitive function in different stages of CI. Second, we analyzed these relationships among different stages and types of CI. Finally, we explored the association between sUA and amyloid/tangle/neurodegeneration (ATN) biomarkers. Results We recruited 427 participants from the CADS, including 382 participants with mini-mental state examination (MMSE) evaluation. The levels of sUA were positively correlated with MMSE scores (p < 0.001), and the correlation was prominent in the course of dementia and in the type of Alzheimer’s disease (AD). The levels of UA had a positive correlation with plasma amyloid-β 42 (Aβ42) (p = 0.004). Higher levels of sUA weakened the correlation of MMSE scores with CSF ATN biomarkers and the correlation of CSF Aβ42 with tau. Conclusion UA is positively correlated with cognitive function, especially in the advanced stage of AD. The probable neuroprotective effects of sUA mainly act on Aβ42 and the downstream pathological cascade.
Collapse
Affiliation(s)
- Shan Huang
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jun Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Dong-Yu Fan
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Tong Luo
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Yanli Li
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, China
| | - Yun-Feng Tu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- Department of Biomedical Engineering, Chongqing University, Chongqing, China
| | - Ying-Ying Shen
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Dong-Wan Chen
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Ye-Ran Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Li-Yong Chen
- Department of Anaesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences (CAS), Shanghai, China
- *Correspondence: Yan-Jiang Wang,
| | - Junhong Guo
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, China
- Junhong Guo,
| |
Collapse
|
48
|
Orellana A, García-González P, Valero S, Montrreal L, de Rojas I, Hernández I, Rosende-Roca M, Vargas L, Tartari JP, Esteban-De Antonio E, Bojaryn U, Narvaiza L, Alarcón-Martín E, Alegret M, Alcolea D, Lleó A, Tárraga L, Pytel V, Cano A, Marquié M, Boada M, Ruiz A. Establishing In-House Cutoffs of CSF Alzheimer’s Disease Biomarkers for the AT(N) Stratification of the Alzheimer Center Barcelona Cohort. Int J Mol Sci 2022; 23:ijms23136891. [PMID: 35805894 PMCID: PMC9266894 DOI: 10.3390/ijms23136891] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Clinical diagnosis of Alzheimer’s disease (AD) increasingly incorporates CSF biomarkers. However, due to the intrinsic variability of the immunodetection techniques used to measure these biomarkers, establishing in-house cutoffs defining the positivity/negativity of CSF biomarkers is recommended. However, the cutoffs currently published are usually reported by using cross-sectional datasets, not providing evidence about its intrinsic prognostic value when applied to real-world memory clinic cases. Methods: We quantified CSF Aβ1-42, Aβ1-40, t-Tau, and p181Tau with standard INNOTEST® ELISA and Lumipulse G® chemiluminescence enzyme immunoassay (CLEIA) performed on the automated Lumipulse G600II. Determination of cutoffs included patients clinically diagnosed with probable Alzheimer’s disease (AD, n = 37) and subjective cognitive decline subjects (SCD, n = 45), cognitively stable for 3 years and with no evidence of brain amyloidosis in 18F-Florbetaben-labeled positron emission tomography (FBB-PET). To compare both methods, a subset of samples for Aβ1-42 (n = 519), t-Tau (n = 399), p181Tau (n = 77), and Aβ1-40 (n = 44) was analyzed. Kappa agreement of single biomarkers and Aβ1-42/Aβ1-40 was evaluated in an independent group of mild cognitive impairment (MCI) and dementia patients (n = 68). Next, established cutoffs were applied to a large real-world cohort of MCI subjects with follow-up data available (n = 647). Results: Cutoff values of Aβ1-42 and t-Tau were higher for CLEIA than for ELISA and similar for p181Tau. Spearman coefficients ranged between 0.81 for Aβ1-40 and 0.96 for p181TAU. Passing–Bablok analysis showed a systematic and proportional difference for all biomarkers but only systematic for Aβ1-40. Bland–Altman analysis showed an average difference between methods in favor of CLEIA. Kappa agreement for single biomarkers was good but lower for the Aβ1-42/Aβ1-40 ratio. Using the calculated cutoffs, we were able to stratify MCI subjects into four AT(N) categories. Kaplan–Meier analyses of AT(N) categories demonstrated gradual and differential dementia conversion rates (p = 9.815−27). Multivariate Cox proportional hazard models corroborated these findings, demonstrating that the proposed AT(N) classifier has prognostic value. AT(N) categories are only modestly influenced by other known factors associated with disease progression. Conclusions: We established CLEIA and ELISA internal cutoffs to discriminate AD patients from amyloid-negative SCD individuals. The results obtained by both methods are not interchangeable but show good agreement. CLEIA is a good and faster alternative to manual ELISA for providing AT(N) classification of our patients. AT(N) categories have an impact on disease progression. AT(N) classifiers increase the certainty of the MCI prognosis, which can be instrumental in managing real-world MCI subjects.
Collapse
Affiliation(s)
- Adelina Orellana
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Pablo García-González
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Sergi Valero
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Isabel Hernández
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Maitee Rosende-Roca
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Liliana Vargas
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Ester Esteban-De Antonio
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Urszula Bojaryn
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Leire Narvaiza
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Emilio Alarcón-Martín
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Montserrat Alegret
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Daniel Alcolea
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, 08029 Barcelona, Spain
| | - Alberto Lleó
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, 08029 Barcelona, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Amanda Cano
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Marta Marquié
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
- Correspondence:
| |
Collapse
|
49
|
Fan DY, Jian JM, Huang S, Li WW, Shen YY, Wang Z, Zeng GH, Yi X, Jin WS, Liu YH, Zeng F, Bu XL, Chen LY, Mao QX, Xu ZQ, Yu JT, Wang J, Wang YJ. Establishment of combined diagnostic models of Alzheimer's disease in a Chinese cohort: the Chongqing Ageing & Dementia Study (CADS). Transl Psychiatry 2022; 12:252. [PMID: 35710549 PMCID: PMC9203516 DOI: 10.1038/s41398-022-02016-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/18/2022] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers are essential for the accurate diagnosis of Alzheimer's disease (AD), yet their measurement levels vary widely across centers and regions, leaving no uniform cutoff values to date. Diagnostic cutoff values of CSF biomarkers for AD are lacking for the Chinese population. As a member of the Alzheimer's Association Quality Control program for CSF biomarkers, we aimed to establish diagnostic models based on CSF biomarkers and risk factors for AD in a Chinese cohort. A total of 64 AD dementia patients and 105 age- and sex-matched cognitively normal (CN) controls from the Chongqing Ageing & Dementia Study cohort were included. CSF Aβ42, P-tau181, and T-tau levels were measured by ELISA. Combined biomarker models and integrative models with demographic characteristics were established by logistic regression. The cutoff values to distinguish AD from CN were 933 pg/mL for Aβ42, 48.7 pg/mL for P-tau181 and 313 pg/mL for T-tau. The AN model, including Aβ42 and T-tau, had a higher diagnostic accuracy of 89.9%. Integrating age and APOE ε4 status to AN model (the ANA'E model) increased the diagnostic accuracy to 90.5% and improved the model performance. This study established cutoff values of CSF biomarkers and optimal combined models for AD diagnosis in a Chinese cohort.
Collapse
Affiliation(s)
- Dong-Yu Fan
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China ,grid.410570.70000 0004 1760 6682Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Jie-Ming Jian
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Shan Huang
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China ,grid.263452.40000 0004 1798 4018First Clinical Medical College, Shanxi Medical University, Taiyuan, China ,grid.263452.40000 0004 1798 4018Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, China
| | - Wei-Wei Li
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China ,Department of Neurology, Western Theater General Hospital, Chengdu, China
| | - Ying-Ying Shen
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Zhen Wang
- grid.410570.70000 0004 1760 6682Department of Critical Care Medicine, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gui-Hua Zeng
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Xu Yi
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Wang-Sheng Jin
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Yu-Hui Liu
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Fan Zeng
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Xian-Le Bu
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Li-Yong Chen
- grid.410570.70000 0004 1760 6682Department of Anaesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Qing-Xiang Mao
- grid.410570.70000 0004 1760 6682Department of Anaesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhi-Qiang Xu
- grid.410570.70000 0004 1760 6682Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jin-Tai Yu
- grid.8547.e0000 0001 0125 2443Department 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
| | - Jun Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China. .,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China. .,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China. .,State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China. .,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
50
|
Studart-Neto A, Coutinho AM. From clinical phenotype to proteinopathy: molecular neuroimaging in neurodegenerative dementias. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:24-35. [PMID: 35976328 PMCID: PMC9491407 DOI: 10.1590/0004-282x-anp-2022-s138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Neurodegenerative dementias are characterized by the abnormal accumulation of misfolded proteins. However, its diagnostic criteria are still based on the clinical phenotype. The development of biomarkers allowed in vivo detection of pathophysiological processes. This article aims to make a non-systematic review of the use of molecular neuroimaging as a biomarker. Molecular neuroimaging is based on the use of radiotracers for image acquisition. The radiotracer most used in PET is 18F-fluorodeoxyglucose (FDG), with which it is possible to study the regional brain glucose metabolism. The pattern of regional hypometabolism provides neuroanatomical information on the neurodegenerative process, which, in turn, has a good specificity for each type of proteinopathy. FDG is very useful in the differential diagnosis of neurodegenerative dementias through the regional pattern of involvement, including dementia with Lewy bodies and the spectrum of frontotemporal dementia. More recently, radiotracers with specific ligands to some of the pathological proteins have been developed. Pittsburgh compound B (PIB) labeled with 11C and the ligands that use 18F (florbetapir, florbetaben and flutemetamol) are the most used radiotracers for the detection of insoluble β-amyloid peptide in Alzheimer's disease (AD). A first generation of ligands for tau protein has been developed, but it has some affinity for other non-tau protein aggregates. A second generation has the advantage of having a higher affinity for hyperphosphorylated tau protein, including in primary tauopathies.
Collapse
Affiliation(s)
- Adalberto Studart-Neto
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo, SP, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Radiologia e Oncologia, Divisão e Laboratório de Medicina Nuclear (LIM 43), São Paulo, SP, Brazil
| |
Collapse
|