Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
60
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found

      Discriminative Accuracy of Plasma Phospho-tau217 for Alzheimer Disease vs Other Neurodegenerative Disorders

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Key Points

          Question

          What is the discriminative accuracy of plasma phospho-tau217 (P-tau217) for differentiating Alzheimer disease from other neurodegenerative disorders?

          Findings

          In this cross-sectional study that included 1402 participants from 3 selected cohorts, plasma P-tau217 discriminated Alzheimer disease from other neurodegenerative diseases (area under the receiver operating characteristic curve of 0.89 in a neuropathologically defined cohort and 0.96 in a clinically defined cohort), with performance that was significantly better than established Alzheimer disease plasma- and MRI-based biomarkers but not significantly different from key CSF- or PET-based biomarkers.

          Meaning

          Although plasma P-tau217 was able to discriminate Alzheimer disease from other neurodegenerative diseases, further research is needed to validate the findings in unselected and diverse populations, optimize the assay, and determine its potential role in clinical care.

          Abstract

          Importance

          There are limitations in current diagnostic testing approaches for Alzheimer disease (AD).

          Objective

          To examine plasma tau phosphorylated at threonine 217 (P-tau217) as a diagnostic biomarker for AD.

          Design, Setting, and Participants

          Three cross-sectional cohorts: an Arizona-based neuropathology cohort (cohort 1), including 34 participants with AD and 47 without AD (dates of enrollment, May 2007-January 2019); the Swedish BioFINDER-2 cohort (cohort 2), including cognitively unimpaired participants (n = 301) and clinically diagnosed patients with mild cognitive impairment (MCI) (n = 178), AD dementia (n = 121), and other neurodegenerative diseases (n = 99) (April 2017-September 2019); and a Colombian autosomal-dominant AD kindred (cohort 3), including 365 PSEN1 E280A mutation carriers and 257 mutation noncarriers (December 2013-February 2017).

          Exposures

          Plasma P-tau217.

          Main Outcomes and Measures

          Primary outcome was the discriminative accuracy of plasma P-tau217 for AD (clinical or neuropathological diagnosis). Secondary outcome was the association with tau pathology (determined using neuropathology or positron emission tomography [PET]).

          Results

          Mean age was 83.5 (SD, 8.5) years in cohort 1, 69.1 (SD, 10.3) years in cohort 2, and 35.8 (SD, 10.7) years in cohort 3; 38% were women in cohort 1, 51% in cohort 2, and 57% in cohort 3. In cohort 1, antemortem plasma P-tau217 differentiated neuropathologically defined AD from non-AD (area under the curve [AUC], 0.89 [95% CI, 0.81-0.97]) with significantly higher accuracy than plasma P-tau181 and neurofilament light chain (NfL) (AUC range, 0.50-0.72; P < .05). The discriminative accuracy of plasma P-tau217 in cohort 2 for clinical AD dementia vs other neurodegenerative diseases (AUC, 0.96 [95% CI, 0.93-0.98]) was significantly higher than plasma P-tau181, plasma NfL, and MRI measures (AUC range, 0.50-0.81; P < .001) but not significantly different compared with cerebrospinal fluid (CSF) P-tau217, CSF P-tau181, and tau-PET (AUC range, 0.90-0.99; P > .15). In cohort 3, plasma P-tau217 levels were significantly greater among PSEN1 mutation carriers, compared with noncarriers, from approximately 25 years and older, which is 20 years prior to estimated onset of MCI among mutation carriers. Plasma P-tau217 levels correlated with tau tangles in participants with (Spearman ρ = 0.64; P < .001), but not without (Spearman ρ = 0.15; P = .33), β-amyloid plaques in cohort 1. In cohort 2, plasma P-tau217 discriminated abnormal vs normal tau-PET scans (AUC, 0.93 [95% CI, 0.91-0.96]) with significantly higher accuracy than plasma P-tau181, plasma NfL, CSF P-tau181, CSF Aβ42:Aβ40 ratio, and MRI measures (AUC range, 0.67-0.90; P < .05), but its performance was not significantly different compared with CSF P-tau217 (AUC, 0.96; P = .22).

          Conclusions and Relevance

          Among 1402 participants from 3 selected cohorts, plasma P-tau217 discriminated AD from other neurodegenerative diseases, with significantly higher accuracy than established plasma- and MRI-based biomarkers, and its performance was not significantly different from key CSF- or PET-based measures. Further research is needed to optimize the assay, validate the findings in unselected and diverse populations, and determine its potential role in clinical care.

          Abstract

          This cross-sectional study compares the accuracy of plasma tau phosphorylated at threonine 217 (P-tau217) levels vs other plasma-, MRI-, CSF-, and PET-based markers for distinguishing Alzheimer from other neurodegenerative diseases in 3 cohorts in Arizona, Sweden, and Columbia with or at risk for dementia.

          Related collections

          Most cited references34

          • Record: found
          • Abstract: not found
          • Article: not found

          Comparing the Areas under Two or More Correlated Receiver Operating Characteristic Curves: A Nonparametric Approach

            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease

            In 2011, the National Institute on Aging and Alzheimer’s Association created separate diagnostic recommendations for the preclinical, mild cognitive impairment, and dementia stages of Alzheimer’s disease. Scientific progress in the interim led to an initiative by the National Institute on Aging and Alzheimer’s Association to update and unify the 2011 guidelines. This unifying update is labeled a “research framework” because its intended use is for observational and interventional research, not routine clinical care. In the National Institute on Aging and Alzheimer’s Association Research Framework, Alzheimer’s disease (AD) is defined by its underlying pathologic processes that can be documented by postmortem examination or in vivo by biomarkers. The diagnosis is not based on the clinical consequences of the disease (i.e., symptoms/signs) in this research framework, which shifts the definition of AD in living people from a syndromal to a biological construct. The research framework focuses on the diagnosis of AD with biomarkers in living persons. Biomarkers are grouped into those of β amyloid deposition, pathologic tau, and neurodegeneration [AT(N)]. This ATN classification system groups different biomarkers (imaging and biofluids) by the pathologic process each measures. The AT(N) system is flexible in that new biomarkers can be added to the three existing AT(N) groups, and new biomarker groups beyond AT(N) can be added when they become available. We focus on AD as a continuum, and cognitive staging may be accomplished using continuous measures. However, we also outline two different categorical cognitive schemes for staging the severity of cognitive impairment: a scheme using three traditional syndromal categories and a six-stage numeric scheme. It is important to stress that this framework seeks to create a common language with which investigators can generate and test hypotheses about the interactions among different pathologic processes (denoted by biomarkers) and cognitive symptoms. We appreciate the concern that this biomarker-based research framework has the potential to be misused. Therefore, we emphasize, first, it is premature and inappropriate to use this research framework in general medical practice. Second, this research framework should not be used to restrict alternative approaches to hypothesis testing that do not use biomarkers. There will be situations where biomarkers are not available or requiring them would be counterproductive to the specific research goals (discussed in more detail later in the document). Thus, biomarker-based research should not be considered a template for all research into age-related cognitive impairment and dementia; rather, it should be applied when it is fit for the purpose of the specific research goals of a study. Importantly, this framework should be examined in diverse populations. Although it is possible that β-amyloid plaques and neurofibrillary tau deposits are not causal in AD pathogenesis, it is these abnormal protein deposits that define AD as a unique neurodegenerative disease among different disorders that can lead to dementia. We envision that defining AD as a biological construct will enable a more accurate characterization and understanding of the sequence of events that lead to cognitive impairment that is associated with AD, as well as the multifactorial etiology of dementia. This approach also will enable a more precise approach to interventional trials where specific pathways can be targeted in the disease process and in the appropriate people.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Neuropathological stageing of Alzheimer-related changes

              Eighty-three brains obtained at autopsy from nondemented and demented individuals were examined for extracellular amyloid deposits and intraneuronal neurofibrillary changes. The distribution pattern and packing density of amyloid deposits turned out to be of limited significance for differentiation of neuropathological stages. Neurofibrillary changes occurred in the form of neuritic plaques, neurofibrillary tangles and neuropil threads. The distribution of neuritic plaques varied widely not only within architectonic units but also from one individual to another. Neurofibrillary tangles and neuropil threads, in contrast, exhibited a characteristic distribution pattern permitting the differentiation of six stages. The first two stages were characterized by an either mild or severe alteration of the transentorhinal layer Pre-alpha (transentorhinal stages I-II). The two forms of limbic stages (stages III-IV) were marked by a conspicuous affection of layer Pre-alpha in both transentorhinal region and proper entorhinal cortex. In addition, there was mild involvement of the first Ammon's horn sector. The hallmark of the two isocortical stages (stages V-VI) was the destruction of virtually all isocortical association areas. The investigation showed that recognition of the six stages required qualitative evaluation of only a few key preparations.
                Bookmark

                Author and article information

                Journal
                JAMA
                JAMA
                JAMA
                JAMA
                American Medical Association
                0098-7484
                1538-3598
                25 August 2020
                28 July 2020
                28 January 2021
                : 324
                : 8
                : 1-11
                Affiliations
                [1 ]Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
                [2 ]Memory Clinic, Skåne University Hospital, Malmö, Sweden
                [3 ]Massachusetts General Hospital, Harvard Medical School, Boston
                [4 ]Grupo de Neurociencias de Antioquia of Universidad de Antioquia, Medellin, Colombia
                [5 ]Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
                [6 ]Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
                [7 ]Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
                [8 ]UK Dementia Research Institute at UCL, London, United Kingdom
                [9 ]Banner Alzheimer’s Institute, Phoenix, Arizona
                [10 ]Banner Sun Health Research Institute, Sun City, Arizona
                [11 ]Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
                [12 ]Department of Neurology, Skåne University Hospital, Lund, Sweden
                [13 ]Molecular Neuropathology of Alzheimer’s Disease (MoNeA), Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
                [14 ]Eli Lilly and Company, Indianapolis, Indiana
                [15 ]University of Arizona, Phoenix
                [16 ]Arizona State University, Phoenix
                [17 ]Translational Genomics Research Institute, Phoenix, Arizona
                Author notes
                Article Information
                Corresponding Author: Oskar Hansson, MD, PhD, Memory Clinic, Skåne University Hospital, SE-20502 Malmö, Sweden ( Oskar.Hansson@ 123456med.lu.se ).
                Accepted for Publication: June 22, 2020.
                Published Online: July 28, 2020. doi:10.1001/jama.2020.12134
                Author Contributions: Drs Hansson and Palmqvist had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Drs Palmqvist and Janelidze contributed equally as first authors. Drs Hansson and Reiman (nonequally) contributed as senior authors.
                Concept and design: Hansson.
                Acquisition, analysis, or interpretation of data: All authors.
                Drafting of the manuscript: Palmqvist, Janelidze, Hansson.
                Critical revision of the manuscript for important intellectual content: All authors.
                Statistical analysis: Palmqvist, Janelidze, Su, Chen, Strandberg.
                Obtained funding: Palmqvist, Zetterberg, Mattsson-Carlgren, Beach, Blennow, Dage, Reiman, Hansson.
                Administrative, technical, or material support: Palmqvist, Lopera, Strandberg, Smith, Villegas, Sepulveda-Falla, Chai, Beach, Dage, Reiman, Hansson.
                Supervision: Dage, Reiman, Hansson.
                Conflict of Interest Disclosures: Dr Quiroz reported receiving grants from the National Institutes of Health (NIH) and Massachusetts General Hospital. Dr Zetterberg reported receiving grants from The Knut and Alice Wallenberg Foundation, European Research Council, and Swedish Research Council; receiving personal fees from Samumed, Roche Diagnostics, CogRx, Wave, Alzecure, and Biogen; and that he is cofounder of Brain Biomarker Solutions in Gothenburg AB. Dr Lopera reported receiving grants from the NIH and Genentech/Roche/Banner and receiving personal fees from the NIH. Dr Su reported receiving grants from the NIH, the State of Arizona, BrightFocus Foundation, and Alzheimer's Association and receiving personal fees from Green Valley Pharmaceutical LLC. Dr Chai reported a patent to pTau217 assay and its use, antibodies pending. Dr Beach reported receiving grants from the State of Arizona; receiving personal fees from Prothena Biosciences, Vivid Genomics, and Avid Radiopharmaceuticals; and holding stock options with Vivid Genomics. Dr Blennow reported receiving personal fees from Abcam, Axon, Biogen, Lilly, MagQu, Novartis, and Roche Diagnostics and that he is cofounder of Brain Biomarker Solutions in Gothenburg AB, a GU Ventures-based platform company at the University of Gothenburg. Dr Dage reported a patent pending for compounds and methods targeting human tau. Dr Reiman reported receiving grants from National Institute on Aging and the State of Arizona; receiving philanthropic funding from the Banner Alzheimer’s Foundation, Sun Health Foundation, and Roche/Roche Diagnostics; receiving personal fees from Alkahest, Alzheon, Aural Analytics, Denali, Green Valley, MagQ, Takeda/Zinfandel, United Neuroscience; that he has since submission of manuscript become a cofounder of AlzPath, which aims to further develop P-tau217 and fluid biomarkers and advance their use in research, drug development, and clinical settings; holding a patent owned by Banner Health for a strategy to use biomarkers to accelerate evaluation of Alzheimer prevention therapies; and that he is a principal investigator of prevention trials that include research agreements with Genentech/Roche and Novartis/Amgen, PET studies that include research agreements with Avid/Lilly, and several NIH and Foundation-supported research studies. Dr Hansson reported receiving grants from Roche, Biogen, and Pfizer and receiving nonfinancial support from GE Healthcare, AVID Radiopharmaceuticals, and Euroimmun. No other disclosures were reported.
                Funding/Support: Work at the authors’ research centers was supported by the Swedish Research Council, the Knut and Alice Wallenberg Foundation, Region Skåne, the Marianne and Marcus Wallenberg Foundation, the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University , the Swedish Alzheimer Foundation, the Swedish Brain Foundation, The Parkinson Foundation of Sweden, The Parkinson Research Foundation, the Skåne University Hospital Foundation, the Greta and Johan Kock Foundation, the Swedish federal government under the ALF agreement, the NIH Office of the Director, the Alzheimer’s Association, the Massachusetts General Hospital Executive Committee on Research, the National Institute of Neurological Disorders and Stroke (U24 NS072026), National Institute on Aging (P30 AG19610), the Arizona Department of Health Services, the Michael J. Fox Foundation for Parkinson’s Research, Banner Alzheimer’s Foundation, Sun Health Foundation, the State of Arizona, and Eli Lilly and Company. Eli Lilly and Company provided material support for P-tau217 sample analysis and salary for Dr Chai, Mr Proctor, and Dr Dage. The precursor of 18F-flutemetamol was provided by GE Healthcare and the precursor of 18F-RO948 was provided by Roche.
                Role of the Funder/Sponsor: The funders had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication. In addition, Eli Lilly and company had the opportunity to review the manuscript before submission but had no veto power. Dr Hansson made the final decision to submit the manuscript to JAMA for publication.
                Meeting Presentation: This study was presented online at the Alzheimer's Association International Conference; July 28, 2020.
                Article
                PMC7388060 PMC7388060 7388060 joi200077
                10.1001/jama.2020.12134
                7388060
                32722745
                0e0c598f-355d-4d59-ab0a-d5567e5f1f0f
                Copyright 2020 American Medical Association. All Rights Reserved.
                History
                : 13 January 2020
                : 22 June 2020
                Funding
                Funded by: Swedish Research Council
                Funded by: Knut and Alice Wallenberg Foundation
                Funded by: Region Skåne
                Funded by: Marianne and Marcus Wallenberg Foundation
                Funded by: Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University
                Funded by: Swedish Alzheimer Foundation
                Funded by: Swedish Brain Foundation
                Funded by: The Parkinson Foundation of Sweden
                Funded by: The Parkinson Research Foundation
                Funded by: Skåne University Hospital Foundation
                Funded by: Greta and Johan Kock Foundation
                Funded by: Swedish federal government
                Funded by: NIH Office of the Director
                Funded by: Alzheimer’s Association
                Funded by: Massachusetts General Hospital Executive Committee on Research
                Funded by: National Institute of Neurological Disorders and Stroke
                Funded by: National Institute on Aging
                Funded by: Arizona Department of Health Services
                Funded by: Michael J. Fox Foundation for Parkinson’s Research
                Funded by: Banner Alzheimer’s Foundation
                Funded by: Sun Health Foundation
                Funded by: State of Arizona
                Funded by: Eli Lilly and Company
                Categories
                Research
                Research
                Original Investigation
                Online First

                Comments

                Comment on this article