51
|
Bos I, Vos S, Vandenberghe R, Scheltens P, Engelborghs S, Frisoni G, Molinuevo JL, Wallin A, Lleó A, Popp J, Martinez-Lage P, Baird A, Dobson R, Legido-Quigley C, Sleegers K, Van Broeckhoven C, Bertram L, ten Kate M, Barkhof F, Zetterberg H, Lovestone S, Streffer J, Visser PJ. The EMIF-AD Multimodal Biomarker Discovery study: design, methods and cohort characteristics. Alzheimers Res Ther 2018; 10:64. [PMID: 29980228 PMCID: PMC6035398 DOI: 10.1186/s13195-018-0396-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 06/08/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND There is an urgent need for novel, noninvasive biomarkers to diagnose Alzheimer's disease (AD) in the predementia stages and to predict the rate of decline. Therefore, we set up the European Medical Information Framework for Alzheimer's Disease Multimodal Biomarker Discovery (EMIF-AD MBD) study. In this report we describe the design of the study, the methods used and the characteristics of the participants. METHODS Participants were selected from existing prospective multicenter and single-center European studies. Inclusion criteria were having normal cognition (NC) or a diagnosis of mild cognitive impairment (MCI) or AD-type dementia at baseline, age above 50 years, known amyloid-beta (Aβ) status, availability of cognitive test results and at least two of the following materials: plasma, DNA, magnetic resonance imaging (MRI) or cerebrospinal fluid (CSF). Targeted and untargeted metabolomic and proteomic analyses were performed in plasma, and targeted and untargeted proteomics were performed in CSF. Genome-wide SNP genotyping, next-generation sequencing and methylation profiling were conducted in DNA. Visual rating and volumetric measures were assessed on MRI. Baseline characteristics were analyzed using ANOVA or chi-square, rate of decline analyzed by linear mixed modeling. RESULTS We included 1221 individuals (NC n = 492, MCI n = 527, AD-type dementia n = 202) with a mean age of 67.9 (SD 8.3) years. The percentage Aβ+ was 26% in the NC, 58% in the MCI, and 87% in the AD-type dementia groups. Plasma samples were available for 1189 (97%) subjects, DNA samples for 929 (76%) subjects, MRI scans for 862 (71%) subjects and CSF samples for 767 (63%) subjects. For 759 (62%) individuals, clinical follow-up data were available. In each diagnostic group, the APOE ε4 allele was more frequent amongst Aβ+ individuals (p < 0.001). Only in MCI was there a difference in baseline Mini Mental State Examination (MMSE) score between the A groups (p < 0.001). Aβ+ had a faster rate of decline on the MMSE during follow-up in the NC (p < 0.001) and MCI (p < 0.001) groups. CONCLUSIONS The characteristics of this large cohort of elderly subjects at various cognitive stages confirm the central roles of Aβ and APOE ε4 in AD pathogenesis. The results of the multimodal analyses will provide new insights into underlying mechanisms and facilitate the discovery of new diagnostic and prognostic AD biomarkers. All researchers can apply for access to the EMIF-AD MBD data by submitting a research proposal via the EMIF-AD Catalog.
Collapse
Affiliation(s)
- Isabelle Bos
- Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Universiteitssingel 40, Box 34, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Stephanie Vos
- Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Rik Vandenberghe
- University Hospital Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Giovanni Frisoni
- University of Geneva, Geneva, Switzerland
- IRCCS Instituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - José Luis Molinuevo
- Alzheimer’s Disease & Other Cognitive Disorders Unit, Hospital Clínic—IDIBAPS, Barcelona, Spain
- Barcelona Beta Brain Research Center, Fundació Pasqual Maragall, Barcelona, Spain
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Moelndal, Sweden
| | - Alberto Lleó
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julius Popp
- Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Pablo Martinez-Lage
- Center for Research and Advanced Therapies, CITA—Alzheimer Foundation, San Sebastian, Spain
| | | | - Richard Dobson
- King’s College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Farr Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
| | | | - Kristel Sleegers
- Neurodegenerative Brain Diseases Group, VIB—Department of Molecular Genetics, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, VIB—Department of Molecular Genetics, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
- School of Public Health, Imperial College London, London, UK
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Mara ten Kate
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute, London, UK
| | | | - Johannes Streffer
- Experimental Medicine, Janssen Pharmaceutical Companies, Beerse, Belgium
| | - Pieter Jelle Visser
- Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
52
|
Hansson O, Mikulskis A, Fagan AM, Teunissen C, Zetterberg H, Vanderstichele H, Molinuevo JL, Shaw LM, Vandijck M, Verbeek MM, Savage M, Mattsson N, Lewczuk P, Batrla R, Rutz S, Dean RA, Blennow K. The impact of preanalytical variables on measuring cerebrospinal fluid biomarkers for Alzheimer's disease diagnosis: A review. Alzheimers Dement 2018; 14:1313-1333. [DOI: 10.1016/j.jalz.2018.05.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Oskar Hansson
- Department of Neurology; Skåne University Hospital; Lund Sweden
- Memory Clinic; Skåne University Hospital; Malmö Sweden
| | | | - Anne M. Fagan
- Department of Neurology; Washington University School of Medicine; St Louis MO USA
| | | | - Henrik Zetterberg
- UK Dementia Research Institute; London UK
- Department of Molecular Neuroscience; UCL Institute of Neurology; London UK
- Clinical Neurochemistry Laboratory; Sahlgrenska University Hospital; Mölndal Sweden
- Department of Psychiatry and Neurochemistry; Sahlgrenska Academy at the University of Gothenburg; Mölndal Sweden
| | | | - Jose Luis Molinuevo
- BarcelonaBeta Brain Research Center; Pasqual Maragall Foundation; Barcelona Spain
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | | | - Marcel M. Verbeek
- Radboud University Medical Center; Departments of Neurology and Laboratory Medicine; Donders Institute for Brain; Cognition and Behaviour; Nijmegen The Netherlands
| | | | - Niklas Mattsson
- Department of Neurology; Skåne University Hospital; Lund Sweden
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy; Universitätsklinikum Erlangen; Friedrich-Alexander Universität Erlangen-Nürnberg; Germany
- Department of Neurodegeneration Diagnostics; Medical University of Bialystok; Poland
| | | | | | - Robert A. Dean
- Department of Pathology and Laboratory Medicine; Indiana University School of Medicine; Indianapolis IN USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory; Sahlgrenska University Hospital; Mölndal Sweden
- Department of Psychiatry and Neurochemistry; Sahlgrenska Academy at the University of Gothenburg; Mölndal Sweden
| |
Collapse
|
53
|
Bennett DA, Gordon BA. Alzheimer disease biomarkers and synucleinopathy. Neurology 2018; 90:537-538. [PMID: 29467302 DOI: 10.1212/wnl.0000000000005171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- David A Bennett
- From the Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL; and Department of Radiology (B.A.G.), Washington University Medical Center, St. Louis, MO.
| | - Brian A Gordon
- From the Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL; and Department of Radiology (B.A.G.), Washington University Medical Center, St. Louis, MO
| |
Collapse
|
54
|
Trombetta BA, Carlyle BC, Koenig AM, Shaw LM, Trojanowski JQ, Wolk DA, Locascio JJ, Arnold SE. The technical reliability and biotemporal stability of cerebrospinal fluid biomarkers for profiling multiple pathophysiologies in Alzheimer's disease. PLoS One 2018; 13:e0193707. [PMID: 29505610 PMCID: PMC5837100 DOI: 10.1371/journal.pone.0193707] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/19/2018] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) is a complex neurodegenerative disease driven by multiple interacting pathophysiological processes that ultimately results in synaptic loss, neuronal death, and dementia. We implemented a fit-for-purpose modeled approach to qualify a broad selection of commercially available immunoassays and evaluate the biotemporal stability of analytes across five pathophysiological domains of interest in AD, including core amyloid-β (Aβ) and tau AD biomarkers, neurodegeneration, inflammation/immune modulation, neurovascular injury, and metabolism/oxidative stress. METHODS Paired baseline and eight-week CSFs from twenty participants in a clinical drug trial for mild cognitive impairment (MCI) or mild dementia due to AD were used to evaluate sensitivity, intra-assay precision, inter-assay replicability, and eight-week biotemporal stability for sixty unique analytes measured with commercially available single- and multi-plex ELISA assays. Coefficients of variation (CV) were calculated, and intraclass correlation and Wilcoxon signed rank tests were applied. RESULTS We identified 32 biomarker candidates with good to excellent performance characteristics according to assay technical performance and CSF analyte biotemporal stability cut-off criteria. These included: 1) the core AD biomarkers Aβ1-42, Aβ1-40, Aβ1-38, and total tau; 2) non-Aβ, non-tau neurodegeneration markers NfL and FABP3; 3) inflammation/immune modulation markers IL-6, IL-7, IL-8, IL-12/23p40, IL-15, IL-16, MCP-1, MDC, MIP-1β, and YKL-40; 4) neurovascular markers Flt-1, ICAM-1, MMP-1, MMP-2, MMP-3, MMP-10, PlGF, VCAM-1, VEGF, VEGF-C, and VEGF-D; and 5) metabolism/oxidative stress markers 24-OHC, adiponectin, leptin, soluble insulin receptor, and 8-OHdG. CONCLUSIONS Assays for these CSF analytes demonstrate consistent sensitivity, reliability, and biotemporally stability for use in a multiple pathophysiological CSF biomarker panel to profile AD. Their qualification enables further investigation for use in AD diagnosis, staging and progression, disease mechanism profiling, and clinical trials.
Collapse
Affiliation(s)
- Bianca A. Trombetta
- MGH Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Becky C. Carlyle
- MGH Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Aaron M. Koenig
- MGH Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Leslie M. Shaw
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - John Q. Trojanowski
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - David A. Wolk
- Penn Memory Center, Department of Neurology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Joseph J. Locascio
- MGH Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Steven E. Arnold
- MGH Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
- * E-mail:
| |
Collapse
|
55
|
Waldron AM, Wyffels L, Verhaeghe J, Richardson JC, Schmidt M, Stroobants S, Langlois X, Staelens S. Longitudinal Characterization of [18F]-FDG and [18F]-AV45 Uptake in the Double Transgenic TASTPM Mouse Model. J Alzheimers Dis 2018; 55:1537-1548. [PMID: 27911309 PMCID: PMC5181675 DOI: 10.3233/jad-160760] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We aimed to monitor the timing of amyloid-β deposition in relation to changes in brain function using in vivo imaging with [18F]-AV45 and [18F]-FDG in a mouse model of Alzheimer’s disease. TASTPM transgenic mice and wild-type controls were scanned longitudinally with [18F]-AV45 and [18F]-FDG before (3 months of age) and at multiple time points after the onset of amyloid deposition (6, 9, 12, and 15 months of age). As expected with increasing amyloidosis, TASTPM mice demonstrated progressive age-dependent increases in [18F]-AV45 uptake that were significantly higher than for WT from 9 months onwards and correlated to ex vivo measures of amyloid burden. The metabolism of [18F]-AV45 produces several brain penetrant radiometabolites and normalization to a reference region helps to negate this non-specific binding and improve the sensitivity of [18F]-AV45. The observed trajectory of [18F]-FDG alterations deviated from our proposed hypothesis of gradual decreases with worsening amyloidosis. While [18F]-FDG uptake in TASTPM mice was significantly lower than that of WT at 9 months, reduced [18F]-FDG was not associated with aging in TASTPM mice. Moreover, [18F]-FDG uptake did not correlate to measures of ex vivo amyloid burden. Our findings suggest that while amyloid-β is sufficient to induce hypometabolism, these pathologies are not linked in a dose-dependent manner in TASTPM mice.
Collapse
Affiliation(s)
- Ann-Marie Waldron
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.,Department of Nuclear Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | | | - Mark Schmidt
- Department of Neuroscience, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.,Department of Nuclear Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Xavier Langlois
- Department of Neuroscience, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
56
|
Wilson JL, Altman RB. Biomarkers: Delivering on the expectation of molecularly driven, quantitative health. Exp Biol Med (Maywood) 2018; 243:313-322. [PMID: 29199461 PMCID: PMC5813871 DOI: 10.1177/1535370217744775] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Biomarkers are the pillars of precision medicine and are delivering on expectations of molecular, quantitative health. These features have made clinical decisions more precise and personalized, but require a high bar for validation. Biomarkers have improved health outcomes in a few areas such as cancer, pharmacogenetics, and safety. Burgeoning big data research infrastructure, the internet of things, and increased patient participation will accelerate discovery in the many areas that have not yet realized the full potential of biomarkers for precision health. Here we review themes of biomarker discovery, current implementations of biomarkers for precision health, and future opportunities and challenges for biomarker discovery. Impact statement Precision medicine evolved because of the understanding that human disease is molecularly driven and is highly variable across patients. This understanding has made biomarkers, a diverse class of biological measurements, more relevant for disease diagnosis, monitoring, and selection of treatment strategy. Biomarkers' impact on precision medicine can be seen in cancer, pharmacogenomics, and safety. The successes in these cases suggest many more applications for biomarkers and a greater impact for precision medicine across the spectrum of human disease. The authors assess the status of biomarker-guided medical practice by analyzing themes for biomarker discovery, reviewing the impact of these markers in the clinic, and highlight future and ongoing challenges for biomarker discovery. This work is timely and relevant, as the molecular, quantitative approach of precision medicine is spreading to many disease indications.
Collapse
Affiliation(s)
- Jennifer L Wilson
- Bioengineering Department, Stanford University, Stanford, CA 94305, USA
| | - Russ B Altman
- Bioengineering Department, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
57
|
Rodriguez-Vieitez E, Nordberg A. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol 2018; 1750:231-251. [PMID: 29512077 DOI: 10.1007/978-1-4939-7704-8_16] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The recent progress in the development of in vivo biomarkers is rapidly changing how neurodegenerative diseases are conceptualized and diagnosed, and how clinical trials are designed today. Alzheimer's disease (AD)-the most common neurodegenerative disorder-is characterized by a complex neuropathology involving the deposition of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles (NFT) of hyperphosphorylated tau proteins, accompanied by the activation of glial cells-astrocytes and microglia-and neuroinflammatory responses, leading to neurodegeneration and cognitive dysfunction. An increasing diversity of positron emission tomography (PET) imaging radiotracers are available to selectively target the different pathophysiological processes of AD. Along with the success of Aβ PET and the more recent tau PET imaging, there is also a great interest to develop PET tracers to image glial activation and neuroinflammation. While most research to date has focused on imaging microgliosis, recent studies using 11C-deuterium-L-deprenyl (11C-DED) PET imaging suggest that astrocytosis may be present from very early stages of disease development in AD. This chapter provides a detailed description of the practical approach used for the analysis of 11C-DED PET imaging data in a multitracer PET paradigm including 11C-Pittsburgh compound B (11C-PiB) and 18F-fluorodeoxyglucose (18F-FDG). The multitracer PET approach allows investigating the comparative regional and temporal patterns of in vivo brain astrocytosis, fibrillar Aβ deposition, and glucose metabolism in patients at different stages of disease progression. This chapter attempts to stimulate further research in the field, including the development of novel PET tracers that may allow visualizing different aspects of the complex astrocytic and microglial responses in neurodegenerative diseases. Progress in the field will contribute to the incorporation of PET imaging of glial activation and neuroinflammation as biomarkers with clinical application, and motivate further investigation on glial cells as therapeutic targets in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Translational Alzheimer Neurobiology, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Agneta Nordberg
- Division of Translational Alzheimer Neurobiology, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
58
|
|
59
|
La Joie R, Bejanin A, Fagan AM, Ayakta N, Baker SL, Bourakova V, Boxer AL, Cha J, Karydas A, Jerome G, Maass A, Mensing A, Miller ZA, O'Neil JP, Pham J, Rosen HJ, Tsai R, Visani AV, Miller BL, Jagust WJ, Rabinovici GD. Associations between [ 18F]AV1451 tau PET and CSF measures of tau pathology in a clinical sample. Neurology 2017; 90:e282-e290. [PMID: 29282337 DOI: 10.1212/wnl.0000000000004860] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/04/2017] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE To assess the relationships between fluid and imaging biomarkers of tau pathology and compare their diagnostic utility in a clinically heterogeneous sample. METHODS Fifty-three patients (28 with clinical Alzheimer disease [AD] and 25 with non-AD clinical neurodegenerative diagnoses) underwent β-amyloid (Aβ) and tau ([18F]AV1451) PET and lumbar puncture. CSF biomarkers (Aβ42, total tau [t-tau], and phosphorylated tau [p-tau]) were measured by multianalyte immunoassay (AlzBio3). Receiver operator characteristic analyses were performed to compare discrimination of Aβ-positive AD from non-AD conditions across biomarkers. Correlations between CSF biomarkers and PET standardized uptake value ratios (SUVR) were assessed using skipped Pearson correlation coefficients. Voxelwise analyses were run to assess regional CSF-PET associations. RESULTS [18F]AV1451-PET cortical SUVR and p-tau showed excellent discrimination between Aβ-positive AD and non-AD conditions (area under the curve 0.92-0.94; ≤0.83 for other CSF measures), and reached 83% classification agreement. In the full sample, cortical [18F]AV1451 was associated with all CSF biomarkers, most strongly with p-tau (r = 0.75 vs 0.57 for t-tau and -0.49 for Aβ42). When restricted to Aβ-positive patients with AD, [18F]AV1451 SUVR correlated modestly with p-tau and t-tau (both r = 0.46) but not Aβ42 (r = 0.02). On voxelwise analysis, [18F]AV1451 correlated with CSF p-tau in temporoparietal cortices and with t-tau in medial prefrontal regions. Within AD, Mini-Mental State Examination scores were associated with [18F]AV1451-PET, but not CSF biomarkers. CONCLUSION [18F]AV1451-PET and CSF p-tau had comparable value for differential diagnosis. Correlations were robust in a heterogeneous clinical group but attenuated (although significant) in AD, suggesting that fluid and imaging biomarkers capture different aspects of tau pathology. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that, in a clinical sample of patients with a variety of suspected neurodegenerative diseases, both CSF p-tau and [18F]AV1451 distinguish AD from non-AD conditions.
Collapse
Affiliation(s)
- Renaud La Joie
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley.
| | - Alexandre Bejanin
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Anne M Fagan
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Nagehan Ayakta
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Suzanne L Baker
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Viktoriya Bourakova
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Adam L Boxer
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Jungho Cha
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Anna Karydas
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Gina Jerome
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Anne Maass
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Ashley Mensing
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Zachary A Miller
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - James P O'Neil
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Julie Pham
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Howard J Rosen
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Richard Tsai
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Adrienne V Visani
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Bruce L Miller
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - William J Jagust
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| | - Gil D Rabinovici
- From the Memory and Aging Center (R.L.J., A.B., N.A., V.B., A.L.B., J.C., A.K., A.M., Z.A.M., J.P., H.J.R., R.T., A.V., B.L.M., G.D.R.), University of California San Francisco; Knight Alzheimer's Disease Research Center (A.M.F., G.J.), Department of Neurology (A.M.F., G.J.), and The Hope Center for Neurological Disorders (A.M.F., G.J.), Washington University in St. Louis, MO; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., J.P.O., W.J.J.), Lawrence Berkeley National Laboratory, Berkeley, CA; and Helen Wills Neuroscience Institute (A.M., W.J.J., G.D.R.), University of California Berkeley
| |
Collapse
|
60
|
Belathur Suresh M, Fischl B, Salat DH. Factors influencing accuracy of cortical thickness in the diagnosis of Alzheimer's disease. Hum Brain Mapp 2017; 39:1500-1515. [PMID: 29271096 DOI: 10.1002/hbm.23922] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/28/2017] [Accepted: 12/07/2017] [Indexed: 02/04/2023] Open
Abstract
There is great value to use of structural neuroimaging in the assessment of Alzheimer's disease (AD). However, to date, predictive value of structural imaging tend to range between 80% and 90% in accuracy and it is unclear why this is the case given that structural imaging should parallel the pathologic processes of AD. There is a possibility that clinical misdiagnosis relative to the gold standard pathologic diagnosis and/or additional brain pathologies are confounding factors contributing to reduced structural imaging classification accuracy. We examined potential factors contributing to misclassification of individuals with clinically diagnosed AD purely from cortical thickness measures. Correctly classified and incorrectly classified groups were compared across a range of demographic, biological, and neuropsychological data including cerebrospinal fluid biomarkers, amyloid imaging, white matter hyperintensity (WMH) volume, cognitive, and genetic factors. Individual subject analyses suggested that at least a portion of the control individuals misclassified as AD from structural imaging additionally harbor substantial AD biomarker pathology and risk, yet are relatively resistant to cognitive symptoms, likely due to "cognitive reserve," and therefore clinically unimpaired. In contrast, certain clinical control individuals misclassified as AD from cortical thickness had increased WMH volume relative to other controls in the sample, suggesting that vascular conditions may contribute to classification accuracy from cortical thickness measures. These results provide examples of factors that contribute to the accuracy of structural imaging in predicting a clinical diagnosis of AD, and provide important information about considerations for future work aimed at optimizing structural based diagnostic classifiers for AD.
Collapse
Affiliation(s)
- Mahanand Belathur Suresh
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Information Science and Engineering, Sri Jayachamarajendra College of Engineering, Mysuru, Karnataka, India
| | - Bruce Fischl
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - David H Salat
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Neuroimaging Research for Veterans Center, VA Boston Healthcare System, Boston, Massachusetts
| | | |
Collapse
|
61
|
Tau-based therapies in neurodegeneration: opportunities and challenges. Nat Rev Drug Discov 2017; 16:863-883. [DOI: 10.1038/nrd.2017.155] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
62
|
Mattsson N, Andreasson U, Zetterberg H, Blennow K. Association of Plasma Neurofilament Light With Neurodegeneration in Patients With Alzheimer Disease. JAMA Neurol 2017; 74:557-566. [PMID: 28346578 PMCID: PMC5822204 DOI: 10.1001/jamaneurol.2016.6117] [Citation(s) in RCA: 700] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Question What is the importance of plasma neurofilament light in Alzheimer disease? Findings In this case-control study of 193 cognitively healthy controls, 197 patients with mild cognitive impairment, and 180 patients with Alzheimer disease dementia, plasma neurofilament light was associated with Alzheimer disease and correlated with future progression of cognitive decline, brain atrophy, and brain hypometabolism. Meaning Plasma neurofilament light may be a promising noninvasive biomarker for Alzheimer disease. Importance Existing cerebrospinal fluid (CSF) or imaging (tau positron emission tomography) biomarkers for Alzheimer disease (AD) are invasive or expensive. Biomarkers based on standard blood test results would be useful in research, drug development, and clinical practice. Plasma neurofilament light (NFL) has recently been proposed as a blood-based biomarker for neurodegeneration in dementias. Objective To test whether plasma NFL concentrations are increased in AD and associated with cognitive decline, other AD biomarkers, and imaging evidence of neurodegeneration. Design, Setting, and Participants In this prospective case-control study, an ultrasensitive assay was used to measure plasma NFL concentration in 193 cognitively healthy controls, 197 patients with mild cognitive impairment (MCI), and 180 patients with AD dementia from the Alzheimer’s Disease Neuroimaging Initiative. The study dates were September 7, 2005, to February 13, 2012. The plasma NFL analysis was performed in September 2016. Main Outcomes and Measures Associations were tested between plasma NFL and diagnosis, Aβ pathologic features, CSF biomarkers of neuronal injury, cognition, brain structure, and metabolism. Results Among 193 cognitively healthy controls, 197 patients with mild cognitive impairment, and 180 patients with AD with dementia, plasma NFL correlated with CSF NFL (Spearman ρ = 0.59, P < .001). Plasma NFL was increased in patients with MCI (mean, 42.8 ng/L) and patients with AD dementia (mean, 51.0 ng/L) compared with controls (mean, 34.7 ng/L) (P < .001) and had high diagnostic accuracy for patients with AD with dementia vs controls (area under the receiver operating characteristic curve, 0.87, which is comparable to established CSF biomarkers). Plasma NFL was particularly high in patients with MCI and patients with AD dementia with Aβ pathologic features. High plasma NFL correlated with poor cognition and AD-related atrophy (at baseline and longitudinally) and with brain hypometabolism (longitudinally). Conclusions and Relevance Plasma NFL is associated with AD diagnosis and with cognitive, biochemical, and imaging hallmarks of the disease. This finding implies a potential usefulness for plasma NFL as a noninvasive biomarker in AD.
Collapse
Affiliation(s)
- Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden2Memory Clinic, Skåne University Hospital, Scania, Sweden3Department of Neurology, Skåne University Hospital, Scania, Sweden
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden5Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Möndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden5Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Möndal, Sweden6Department of Molecular Neuroscience, University College London Institute of Neurology, Queen Square, London, England
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden5Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Möndal, Sweden
| | | |
Collapse
|
63
|
Alzheimer's disease drug development pipeline: 2017. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:367-384. [PMID: 29067343 PMCID: PMC5651419 DOI: 10.1016/j.trci.2017.05.002] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION There is an urgent need to develop new treatments for Alzheimer's disease (AD) and to understand the drug development process for new AD therapies. METHODS We assessed the agents in the AD pipeline as documented in clinicaltrials.gov for phase I, phase II, and phase III, accessed 1/5/2017. RESULTS There are 105 agents in the AD treatment development pipeline, of which 25 agents are in 29 trials in phase I, 52 agents are in 68 trials in phase II, and 28 agents are in 42 trials in phase III. Seventy percent of drugs in the AD pipeline are disease-modifying therapies (DMTs). Fourteen percent are symptomatic cognitive enhancers, and 13% are symptomatic agents addressing neuropsychiatric and behavioral changes (2% have undisclosed mechanisms). Most trials are sponsored by the biopharmaceutical industry. Trials include patients with preclinical AD (cognitively normal with biomarker evidence of AD), prodromal AD (mild cognitive symptoms and biomarker evidence of AD), and AD dementia. Biomarkers are included in many drug development programs particularly those for DMTs. Thirteen of 46 phase II DMT trials have amyloid imaging as an entry criterion, and 10 of 28 phase III trials incorporate amyloid imaging for diagnosis and entry. A large number of participants are needed for AD clinical trials; in total, 54,073 participants are required for trials spanning preclinical AD to AD dementia. When compared with the 2016 pipeline, there are eight new agents in phase I, 16 in phase II, and five in phase III. DISCUSSION The AD drug development pipeline has 105 agents divided among phase I, phase II, and phase III. The trials include a wide range of clinical trial populations, many mechanisms of action, and require a substantial number of clinical trial participants. Biomarkers are increasingly used in patient identification and as outcome measures, particularly in trials of DMTs.
Collapse
|
64
|
Huynh RA, Mohan C. Alzheimer's Disease: Biomarkers in the Genome, Blood, and Cerebrospinal Fluid. Front Neurol 2017; 8:102. [PMID: 28373857 PMCID: PMC5357660 DOI: 10.3389/fneur.2017.00102] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/01/2017] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that slowly destroys memory and thinking skills, resulting in behavioral changes. It is estimated that nearly 36 million are affected globally with numbers reaching 115 million by 2050. AD can only be definitively diagnosed at autopsy since its manifestations of senile plaques and neurofibrillary tangles throughout the brain cannot yet be fully captured with current imaging technologies. Current AD therapeutics have also been suboptimal. Besides identifying markers that distinguish AD from controls, there has been a recent drive to identify better biomarkers that can predict the rates of cognitive decline and neocortical amyloid burden in those who exhibit preclinical, prodromal, or clinical AD. This review covers biomarkers of three main types: genes, cerebrospinal fluid-derived, and blood-derived biomarkers. Looking ahead, cutting-edge OMICs technologies, including proteomics and metabolomics, ought to be fully tapped in order to mine even better biomarkers for AD that are more predictive.
Collapse
Affiliation(s)
- Rose Ann Huynh
- Department of Biomedical Engineering, University of Houston , Houston, TX , USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston , Houston, TX , USA
| |
Collapse
|
65
|
Zhu H, Stern RA, Tao Q, Bourlas A, Essis MD, Chivukula M, Rosenzweig J, Steenkamp D, Xia W, Mercier GA, Tripodis Y, Farlow M, Kowall N, Qiu WQ. An amylin analog used as a challenge test for Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:33-43. [PMID: 28503657 PMCID: PMC5424531 DOI: 10.1016/j.trci.2016.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Preclinical studies demonstrate the potential of amylin in the diagnosis of Alzheimer's disease (AD). We aimed to lay the foundation for repurposing the amylin analog and a diabetes drug, pramlintide, for AD in humans. METHODS We administered a single subcutaneous injection of 60 μg of pramlintide to nondiabetic subjects under fasting conditions. RESULTS None of the participants developed hypoglycemia after the injection of pramlintide. The pramlintide challenge induced a significant surge of amyloid-β peptide and a decrease in total tau in the plasma of AD subjects but not in control participants. The pramlintide injection provoked an increase in interleukin 1 receptor antagonist and a decrease in retinol-binding protein 4, which separates AD subjects from control subjects. DISCUSSION Pramlintide use appeared to be safe in the absence of diabetes. The biomarker changes as a result of the pramlintide challenge, which distinguished AD from control subjects and mild cognitive impairment.
Collapse
Affiliation(s)
- Haihao Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Robert A Stern
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Alexandra Bourlas
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Maritza D Essis
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Meenakshi Chivukula
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - James Rosenzweig
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Devin Steenkamp
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Weiming Xia
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Gustavo A Mercier
- Department of Radiology, Boston University School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Martin Farlow
- Alzheimer's Disease Center, Indiana University, Indianapolis, IN, USA
| | - Neil Kowall
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA.,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
66
|
O'Bryant SE, Mielke MM, Rissman RA, Lista S, Vanderstichele H, Zetterberg H, Lewczuk P, Posner H, Hall J, Johnson L, Fong YL, Luthman J, Jeromin A, Batrla-Utermann R, Villarreal A, Britton G, Snyder PJ, Henriksen K, Grammas P, Gupta V, Martins R, Hampel H. Blood-based biomarkers in Alzheimer disease: Current state of the science and a novel collaborative paradigm for advancing from discovery to clinic. Alzheimers Dement 2017; 13:45-58. [PMID: 27870940 PMCID: PMC5218961 DOI: 10.1016/j.jalz.2016.09.014] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/27/2016] [Indexed: 11/25/2022]
Abstract
The last decade has seen a substantial increase in research focused on the identification of blood-based biomarkers that have utility in Alzheimer's disease (AD). Blood-based biomarkers have significant advantages of being time- and cost-efficient as well as reduced invasiveness and increased patient acceptance. Despite these advantages and increased research efforts, the field has been hampered by lack of reproducibility and an unclear path for moving basic discovery toward clinical utilization. Here we reviewed the recent literature on blood-based biomarkers in AD to provide a current state of the art. In addition, a collaborative model is proposed that leverages academic and industry strengths to facilitate the field in moving past discovery only work and toward clinical use. Key resources are provided. This new public-private partnership model is intended to circumvent the traditional handoff model and provide a clear and useful paradigm for the advancement of biomarker science in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sid E O'Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA.
| | - Michelle M Mielke
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Robert A Rissman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, UCSD School of Medicine, La Jolla, CA, USA
| | - Simone Lista
- AXA Research Fund and UPMC Chair, Paris, France; Department de Neurologie, Institut de la Memorie et de la Maladie d'Alzheimer (IM2A) et Institut du Cerveau et du la Moelle epiniere (ICM), Hospital de la Pitie-Salpetriere, Sorbonne Universites, Universite Pierre et Marie Curie, Paris, France
| | | | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gotenburg, Molndal, Sweden; UCL Institute of Neurology, London, UK
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | | | - James Hall
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Yiu-Lian Fong
- Johnson & Johnson, London Innovation Center, London, UK
| | - Johan Luthman
- Neuroscience Clinical Development, Clinical Neuroscience Eisai, Woodcliff Lake, NJ, USA
| | | | | | - Alcibiades Villarreal
- Centro de Neurociencias y Unidad de Investigacion Clinica, Instituto de Investigaciones Cientificas y Servicios de Alta Tecnologia (INDICASAT AIP), Ciudad del Saber, Panama, Panama
| | - Gabrielle Britton
- Centro de Neurociencias y Unidad de Investigacion Clinica, Instituto de Investigaciones Cientificas y Servicios de Alta Tecnologia (INDICASAT AIP), Ciudad del Saber, Panama, Panama
| | - Peter J Snyder
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Kim Henriksen
- Neurodegenerative Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Paula Grammas
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, RI, USA
| | - Veer Gupta
- Faculty of Health, Engineering and Sciences, Center of Excellence for Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Ralph Martins
- Faculty of Health, Engineering and Sciences, Center of Excellence for Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Harald Hampel
- AXA Research Fund and UPMC Chair, Paris, France; Department de Neurologie, Institut de la Memorie et de la Maladie d'Alzheimer (IM2A) et Institut du Cerveau et du la Moelle epiniere (ICM), Hospital de la Pitie-Salpetriere, Sorbonne Universites, Universite Pierre et Marie Curie, Paris, France
| |
Collapse
|
67
|
Liang K, Zhang J, Yin C, Zhou X, Zhou S. Protective effects and mechanism of TPX2 on neurocyte apoptosis of rats in Alzheimer's disease model. Exp Ther Med 2016; 13:576-580. [PMID: 28352333 PMCID: PMC5348683 DOI: 10.3892/etm.2016.4006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022] Open
Abstract
We investigated the protective effects and mechanism of TPX2 on apoptosis of rat neurocytes. A total of 90 SD rats were randomly divided into the drug group, the control group and the blank group, with 30 rats in each group. The rats in the drug group and in the blank group were anesthetized with 10% chloral hydrate (at the dose of 0.5 ml/100 g) and Aβ1-42, with the concentration of 5 µl (1 µg/µl), was injected in the exact position of bilateral hippocampal areas of rats to establish the model. The configured TPX2 inhibitors and edible benne oil were mixed and made into a suspension. After model establishment, the rats were given different treatment methods; the rats in the drug group were given gavage administration in the proportion of 75 mg/kg once a day. The rats in the control group were given intragastric administration with the same proportion of physiological saline once a day. The blank group was the normal healthy group and the rats in this group did not undergo any surgery or drug treatment. Brain tissue in rats were divided into two parts, one part was fixed, dehydrated, paraffin-embedded and made into slices of approximately 5 µm. TUNEL staining was used to examine the apoptosis of brain tissue, H&E staining was used to observe the brain tissue cells of each group, and western blotting for detecting the MAPK, Erk and expression levels of p38 and RT-polymerase chain reaction method was employed to examine mRNA expression levels of MAPK, Erk and p21. After one week, TUNEL staining showed that apoptosis of brain tissue in the drug group was significantly greater than those of the control and blank groups. The protein expression levels of MAPK, Erk and p38 were significantly higher than those of the control group and the normal healthy group; the differences were statistically significant (P<0.05). Western blotting showed that the protein expression levels of MAPK, Erk and p38 of the drug group were significantly lower than those of the control group but higher than those of the normal healthy group; the differences were statistically significant (P<0.05). TPX2 has a protective effect on the apoptosis of brain tissue processed by Aβ1-42, which plays its role through the inhibition of the protein expression levels of MAPK, Erk and p38.
Collapse
Affiliation(s)
- Keshan Liang
- Department of Neurology, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Neurology, Pingyi Branch of Qilu Hospital, Shandong University, Pingyi, Shandong 273300, P.R. China
| | - Jingling Zhang
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Chengbin Yin
- Department of Emergency, Qingdao Branch of Qilu Hospital of Shandong University, Qingdao, Shandong 266000, P.R. China
| | - Xueying Zhou
- Department of Neurology, Shangdong University of Traditional Chinese Medicine, Jinan, Shandong 250031, P.R. China
| | - Shengnian Zhou
- Department of Neurology, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
68
|
Alosco ML, Tripodis Y, Jarnagin J, Baugh CM, Martin B, Chaisson CE, Estochen N, Song L, Cantu RC, Jeromin A, Stern RA. Repetitive head impact exposure and later-life plasma total tau in former National Football League players. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2016; 7:33-40. [PMID: 28229128 PMCID: PMC5312499 DOI: 10.1016/j.dadm.2016.11.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction Blood protein analysis of total tau (t-tau) may be a practical screening biomarker for chronic traumatic encephalopathy (CTE), a neurodegenerative tauopathy associated with repetitive head impact (RHI) exposure. We examined plasma t-tau in symptomatic former National Football League (NFL) players compared with controls and the relationship between RHI exposure and later-life plasma t-tau. Methods Ninety-six former NFL players (age 40–69) and 25 same-age controls underwent blood draw to determine plasma t-tau levels. The cumulative head impact index (CHII) quantified RHI exposure. Subjects completed measures of clinical function. Results A higher CHII predicted greater plasma t-tau in the former NFL players (P = .0137). No group differences in plasma t-tau emerged, but a concentration ≥3.56 pg/mL was 100% specific to former NFL players. Plasma t-tau did not predict clinical function. Discussion Greater RHI exposure predicted higher later-life plasma t-tau concentrations, and further study on plasma t-tau as a candidate screening biomarker for CTE is warranted.
Collapse
Affiliation(s)
- Michael L Alosco
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Johnny Jarnagin
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA
| | - Christine M Baugh
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Interfaculty Initiative in Health Policy, Harvard University Boston, MA, USA
| | - Brett Martin
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA; Data Coordinating Center, Boston University School of Public Health, Boston, MA, USA
| | - Christine E Chaisson
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA; Data Coordinating Center, Boston University School of Public Health, Boston, MA, USA
| | | | - Linan Song
- Quanterix Corporation, Lexington, MA, USA
| | - Robert C Cantu
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA
| | | | - Robert A Stern
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA; Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
69
|
Mattsson N, Insel PS, Palmqvist S, Portelius E, Zetterberg H, Weiner M, Blennow K, Hansson O. Cerebrospinal fluid tau, neurogranin, and neurofilament light in Alzheimer's disease. EMBO Mol Med 2016; 8:1184-1196. [PMID: 27534871 PMCID: PMC5048367 DOI: 10.15252/emmm.201606540] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cerebrospinal fluid (CSF) tau (total tau, T‐tau), neurofilament light (NFL), and neurogranin (Ng) are potential biomarkers for neurodegeneration in Alzheimer's disease (AD). It is unknown whether these biomarkers provide similar or complementary information in AD. We examined 93 patients with AD, 187 patients with mild cognitive impairment, and 109 controls. T‐tau, Ng, and NFL were all predictors of AD diagnosis. Combinations improved the diagnostic accuracy (AUC 85.5% for T‐tau, Ng, and NFL) compared to individual biomarkers (T‐tau 80.8%; Ng 71.4%; NFL 77.7%). T‐tau and Ng were highly correlated (ρ = 0.79, P < 0.001) and strongly associated with β‐amyloid (Aβ) pathology, and with longitudinal deterioration in cognition and brain structure, primarily in people with Aβ pathology. NFL on the other hand was not associated with Aβ pathology and was associated with cognitive decline and brain atrophy independent of Aβ. T‐tau, Ng, and NFL provide partly independent information about neuronal injury and may be combined to improve the diagnostic accuracy for AD. T‐tau and Ng reflect Aβ‐dependent neurodegeneration, while NFL reflects neurodegeneration independently of Aβ pathology.
Collapse
Affiliation(s)
- Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Philip S Insel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Erik Portelius
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Michael Weiner
- Department of Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Neurology, Skåne University Hospital, Lund, Sweden
| | | |
Collapse
|
70
|
Mattsson N, Zetterberg H, Janelidze S, Insel PS, Andreasson U, Stomrud E, Palmqvist S, Baker D, Tan Hehir CA, Jeromin A, Hanlon D, Song L, Shaw LM, Trojanowski JQ, Weiner MW, Hansson O, Blennow K. Plasma tau in Alzheimer disease. Neurology 2016; 87:1827-1835. [PMID: 27694257 PMCID: PMC5089525 DOI: 10.1212/wnl.0000000000003246] [Citation(s) in RCA: 387] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 07/12/2016] [Indexed: 01/10/2023] Open
Abstract
Objective: To test whether plasma tau is altered in Alzheimer disease (AD) and whether it is related to changes in cognition, CSF biomarkers of AD pathology (including β-amyloid [Aβ] and tau), brain atrophy, and brain metabolism. Methods: This was a study of plasma tau in prospectively followed patients with AD (n = 179), patients with mild cognitive impairment (n = 195), and cognitive healthy controls (n = 189) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and cross-sectionally studied patients with AD (n = 61), mild cognitive impairment (n = 212), and subjective cognitive decline (n = 174) and controls (n = 274) from the Biomarkers for Identifying Neurodegenerative Disorders Early and Reliably (BioFINDER) study at Lund University, Sweden. A total of 1284 participants were studied. Associations were tested between plasma tau and diagnosis, CSF biomarkers, MRI measures, 18fluorodeoxyglucose-PET, and cognition. Results: Higher plasma tau was associated with AD dementia, higher CSF tau, and lower CSF Aβ42, but the correlations were weak and differed between ADNI and BioFINDER. Longitudinal analysis in ADNI showed significant associations between plasma tau and worse cognition, more atrophy, and more hypometabolism during follow-up. Conclusions: Plasma tau partly reflects AD pathology, but the overlap between normal aging and AD is large, especially in patients without dementia. Despite group-level differences, these results do not support plasma tau as an AD biomarker in individual people. Future studies may test longitudinal plasma tau measurements in AD.
Collapse
Affiliation(s)
- Niklas Mattsson
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco.
| | - Henrik Zetterberg
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Shorena Janelidze
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Philip S Insel
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Ulf Andreasson
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Erik Stomrud
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - David Baker
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Cristina A Tan Hehir
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Andreas Jeromin
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - David Hanlon
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Linan Song
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Leslie M Shaw
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - John Q Trojanowski
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Michael W Weiner
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Oskar Hansson
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | - Kaj Blennow
- From the Clinical Memory Research Unit (N.M., S.J., P.S.I., E.S., S.P., O.H.), Department of Clinical Sciences, Malmö, Lund University; Department of Neurology (N.M., E.S., O.H., S.P.), Skåne University Hospital, Lund; Clinical Neurochemistry Laboratory (H.Z., U.A., K.B.), Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Canpus, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK; Janssen R&D (D.B.), Titusville, NJ; Diagnostics and Life Sciences (C.A.T.H.), GE Global Research, Niskayuna, NY; Quanterix Corporation (A.J., D.H., L.S.), Lexington, MA; Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Center for Imaging of Neurodegenerative Diseases (M.W.W.), Department of Veterans Affairs Medical Center; and Department of Radiology and Biomedical Imaging (M.W.W.), University of California, San Francisco
| | | |
Collapse
|
71
|
Bolognesi ML, Gandini A, Prati F, Uliassi E. From Companion Diagnostics to Theranostics: A New Avenue for Alzheimer’s Disease? J Med Chem 2016; 59:7759-70. [DOI: 10.1021/acs.jmedchem.6b00151] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Maria Laura Bolognesi
- Department
of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Annachiara Gandini
- Department
of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136 Trieste, Italy
| | - Federica Prati
- Department
of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- College of Life Sciences,
Sir James Black Centre, University of Dundee, Dundee DD1 5EH, U.K
| | - Elisa Uliassi
- Department
of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| |
Collapse
|
72
|
Rojas C. G, de Guevara DL, Jaimovich F. R, Brunetti E, Faure L. E, Gálvez M. M. NEUROIMÁGENES EN DEMENCIAS. REVISTA MÉDICA CLÍNICA LAS CONDES 2016. [DOI: 10.1016/j.rmclc.2016.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|