1
|
Santos LE, Mattos P, Pinheiro TL, Silva A, Drummond C, Sudo FK, Barros-Aragão F, Vanderborght B, Brandão CO, Ferreira ST, Tovar-Moll F, De Felice FG. Performance of plasma biomarkers for diagnosis and prediction of dementia in a Brazilian cohort. Nat Commun 2025; 16:2911. [PMID: 40133253 PMCID: PMC11937383 DOI: 10.1038/s41467-025-56756-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 01/28/2025] [Indexed: 03/27/2025] Open
Abstract
Despite remarkable progress in the biomarker field in recent years, local validation of plasma biomarkers of Alzheimer's disease (AD) and dementia is still lacking in Latin America. In this longitudinal cohort study of 145 elderly Brazilians, we assess the diagnostic performance of plasma biomarkers, based on clinical diagnosis and CSF biomarker positivity. Follow-up data of up to 4.7 years were used to determine performance in predicting diagnostic conversions. Participants were clinically categorized as cognitively unimpaired (n = 49), amnestic mild cognitive impairment (n = 29), AD (n = 38), Lewy body dementia (n = 22), or vascular dementia (n = 7). Plasma Tau, Aβ40, Aβ42, NfL, GFAP, pTau231, pTau181 and pTau217 were measured on the SIMOA HD-X platform. Plasma pTau217 showed excellent performance determining CSF biomarker status in the cohort, either alone (ROC AUC = 0.94, 95% CI: [0.88-1.00]) or as a ratio to Aβ42 (ROC AUC = 0.98, 95% CI: [0.94-1.00]). This study comprises an initial step towards local validation and adoption of dementia biomarkers in Brazil.
Collapse
Affiliation(s)
- Luis E Santos
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Paulo Mattos
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Program in Morphological Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thais L Pinheiro
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Ananssa Silva
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Claudia Drummond
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Department of Speech and Hearing Pathology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe Kenji Sudo
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | | | - Bart Vanderborght
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | | | - Sergio T Ferreira
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Fernanda G De Felice
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences and Department of Psychiatry, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
2
|
Schinke H, Förnvik Jonsson M, Gummesson M, Nilsson R, Gaupp S, Manuilova E, McIlwrick S, Weinberger JP, Rutz S, Carboni M, Stomrud E. Concordance between the updated Elecsys cerebrospinal fluid immunoassays and amyloid positron emission tomography for Alzheimer's disease assessment: findings from the Apollo study. Clin Chem Lab Med 2025:cclm-2024-1476. [PMID: 40068924 DOI: 10.1515/cclm-2024-1476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 04/13/2025]
Abstract
OBJECTIVES The Apollo study was designed to support the clinical performance verification of the adjusted cutoffs of the Elecsys® β-Amyloid(1-42) (Aβ42) cerebrospinal fluid (CSF) II, β-Amyloid(1-40) (Aβ40) CSF, Phospho-Tau (181P) (pTau) CSF and Total-Tau (tTau) CSF immunoassays (Roche Diagnostics International Ltd) for measuring fresh CSF samples, and assess the concordance of the Elecsys CSF pTau/Aβ42, tTau/Aβ42 and Aβ42/Aβ40 ratios, as well as Aβ42 alone, with amyloid positron emission tomography (PET) visual read status. METHODS The primary study endpoint was to assess the concordance of the Elecsys CSF ratios and Aβ42 alone with amyloid PET visual read status using fresh CSF samples collected from individuals with subjective cognitive decline or mild cognitive impairment, handled with a new routine-use pre-analytical procedure and measured with the Elecsys CSF immunoassays. The sample stability after 1- to 13-week storage at -20 °C was also investigated in an exploratory analysis. RESULTS Of 108 screened individuals, 91 met the eligibility criteria, of whom 44.0 % were amyloid PET-positive and 56.0 % amyloid PET-negative. Positive percent agreement (PPA) and negative percent agreement, respectively, were 0.800 and 0.882 for pTau/Aβ42, 0.775 and 0.902 for tTau/Aβ42, and 0.950 and 0.824 for Aβ42/Aβ40. For Aβ42, PPA was 0.975 and negative likelihood ratio was 0.039. Overall, 33 samples (36.3 %) were frozen at -20 °C for 1-13 weeks. All concentration recoveries were within 100 ± 10 % when stored at -20 °C for ≤8 weeks. CONCLUSIONS Elecsys CSF ratios and Aβ42 alone may be reliable alternatives to amyloid PET for identifying amyloid positivity in clinical practice.
Collapse
Affiliation(s)
| | - Magnus Förnvik Jonsson
- Department of Clinical Chemistry and Pharmacology, Skåne University Hospital, Lund, Sweden
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Mayme Gummesson
- Department of Clinical Chemistry and Pharmacology, Skåne University Hospital, Lund, Sweden
| | - Rikard Nilsson
- Department of Clinical Chemistry and Pharmacology, Skåne University Hospital, Lund, Sweden
| | | | | | | | | | | | | | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
3
|
Leuzy A, Bollack A, Pellegrino D, Teunissen CE, La Joie R, Rabinovici GD, Franzmeier N, Johnson K, Barkhof F, Shaw LM, Arkhipenko A, Schindler SE, Honig LS, Moscoso Rial A, Schöll M, Zetterberg H, Blennow K, Hansson O, Farrar G. Considerations in the clinical use of amyloid PET and CSF biomarkers for Alzheimer's disease. Alzheimers Dement 2025; 21:e14528. [PMID: 40042435 PMCID: PMC11881640 DOI: 10.1002/alz.14528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 03/09/2025]
Abstract
Amyloid-β (Aβ) positron emission tomography (PET) imaging and cerebrospinal fluid (CSF) biomarkers are now established tools in the diagnostic workup of patients with Alzheimer's disease (AD), and their use is anticipated to increase with the introduction of new disease-modifying therapies. Although these biomarkers are comparable alternatives in research settings to determine Aβ status, biomarker testing in clinical practice requires careful consideration of the strengths and limitations of each modality, as well as the specific clinical context, to identify which test is best suited for each patient. This article provides a comprehensive review of the pathologic processes reflected by Aβ-PET and CSF biomarkers, their performance, and their current and future applications and contexts of use. The primary aim is to assist clinicians in making better-informed decisions about the suitability of each biomarker in different clinical situations, thereby reducing the risk of misdiagnosis or incorrect interpretation of biomarker results. HIGHLIGHTS: Recent advances have positioned Aβ PET and CSF biomarkers as pivotal in AD diagnosis. It is crucial to understand the differences in the clinical use of these biomarkers. A team of experts reviewed the state of Aβ PET and CSF markers in clinical settings. Differential features in the clinical application of these biomarkers were reviewed. We discussed the role of Aβ PET and CSF in the context of novel plasma biomarkers.
Collapse
Grants
- AF-930351 Neurodegenerative Disease Research
- 101053962 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- R01 AG066107 NIA NIH HHS
- FO2022-0270 Bluefield Project, Olav Thon Foundation, Erling-Persson Family Foundation
- 101112145 European Union's Horizon Europe
- Alzheimer Netherlands
- ZEN-21-848495 Alzheimer's Association 2021 Zenith Award
- 2022-0231 Knut and Alice Wallenberg foundation
- KAW 2023.0371 Knut and Alice Wallenberg Foundation
- U19 ADNI4 Harvard Aging Brain Study
- R01 AG081394 NIA NIH HHS
- ADRC P30-AG-072979 Harvard Aging Brain Study
- 2022-1259 Regionalt Forskningsstöd
- Shanendoah Foundation
- 2020-O000028 Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, Skåne University Hospital Foundation
- The Selfridges Group Foundation
- R56 AG057195 NIA NIH HHS
- U01 NS100600 NINDS NIH HHS
- ALZ2022-0006 Hjärnfonden, Sweden
- U01 AG057195 NIA NIH HHS
- Dutch National Dementia Strategy
- ZEN24-1069572 Alzheimer's Association
- R01AG072474 Harvard Aging Brain Study
- 860197 Marie Curie International Training Network
- AF-939721 Neurodegenerative Disease Research
- R01 AG070941 NIA NIH HHS
- P01 AG036694 NIA NIH HHS
- JPND2021-00694 Neurodegenerative Disease Research
- ADSF-21-831376-C AD Strategic Fund, and Alzheimer's Association
- AF-994900 Swedish Alzheimer Foundation
- NIH
- ALFGBG-813971 County Councils, the ALF-agreement
- FO2021-0293 Swedish Brain Foundation
- U19AG063893 NINDS NIH HHS
- 2022-01018 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- 201809-2016862 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- 831434 Innovative Medicines Initiatives 3TR
- 101132933 European Union's Horizon Europe
- European Union Joint Programme
- Cure Alzheimer's fund, Rönström Family Foundation
- ID 390857198 Munich Cluster for Systems Neurology
- U01-AG057195 NIA NIH HHS
- Deutsche Forschungsgemeinschaft
- 2021-06545 Swedish Research Council
- Sahlgrenska Academy at the University of Gothenburg
- U19 AG024904 NIA NIH HHS
- GE Healthcare
- JPND2019-466-236 European Union Joint Program for Neurodegenerative Disorders
- P30 AG062422 NIA NIH HHS
- ADG-101096455 European Research Council
- 2022-00732 Neurodegenerative Disease Research
- 860197 Marie Skłodowska-Curie
- P01 AG019724 NIA NIH HHS
- U01NS100600 NINDS NIH HHS
- AF-980907 Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson's disease) at Lund University, Swedish Alzheimer Foundation
- P30 AG066462 NIA NIH HHS
- 2022-00775 GHR Foundation, Swedish Research Council
- R44 AG071388 NIA NIH HHS
- FO2017-0243 Hjärnfonden, Sweden
- AF-968270 Neurodegenerative Disease Research
- KAW2014.0363 Knut and Alice Wallenberg Foundation
- SG-23-1061717 Alzheimer's Association
- 2021-02678 Swedish Research Council
- R01 AG059013 NIA NIH HHS
- R35 AG072362 NIA NIH HHS
- VGFOUREG-995510 Västra Götaland Region R&D
- American College of Radiology
- R01 AG081394-01 European Union's Horizon Europe
- R21 AG070768 NIA NIH HHS
- U19 AG063893 NIA NIH HHS
- 2022-Projekt0080 Swedish Federal Government under the ALF agreement
- ALFGBG-965326 County Councils, the ALF-agreement
- Alzheimer Drug Discovery Foundation
- Rainwater Charitable Foundation
- Research of the European Commission
- R01AG083740 National Institute of Aging
- ADSF-21-831381-C AD Strategic Fund, and Alzheimer's Association
- SG-23-1038904 Alzheimer's Association 2022-2025
- RS-2023-00263612 National Research Foundation of Korea
- P30-AG062422 NIA NIH HHS
- R21AG070768 Harvard Aging Brain Study
- 2017-02869 Swedish Research Council
- 101034344 Joint Undertaking
- ALFGBG-715986 Swedish state under the agreement between the Swedish government and the County Councils, ALF-agreement
- ERAPERMED2021-184 ERA PerMed
- U19AG024904 Harvard Aging Brain Study
- R01 AG072474 NIA NIH HHS
- UKDRI-1003 Neurodegenerative Disease Research
- 10510032120003 Health Holland, the Dutch Research Council
- 2019-02397 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- EXC 2145 SyNergy Munich Cluster for Systems Neurology
- 1412/22 Parkinson foundation of Sweden
- R01 AG046396 NIA NIH HHS
- ALFGBG-71320 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- P01-AG019724 NIA NIH HHS
- ALFGBG-965240 Swedish state under the agreement between the Swedish government and the County Councils, ALF-agreement
- Deutsche Parkinson Gesellschaft
- ADSF-21-831377-C AD Strategic Fund, and Alzheimer's Association
- National MS Society
- R01 AG083740 NIA NIH HHS
- 2017-00915 Neurodegenerative Disease Research
- 2023-06188 Swedish Research Council
- Alzheimer Association
- National MS Society
- Alzheimer Netherlands
- NIH
- NIA
- National Institute of Neurological Disorders and Stroke
- American College of Radiology
- Rainwater Charitable Foundation
- Deutsche Forschungsgemeinschaft
- NINDS
- Knut and Alice Wallenberg Foundation
- Swedish Research Council
- National Research Foundation of Korea
- Swedish Brain Foundation
- European Research Council
- Alzheimer's Association
- GE Healthcare
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Department of NeuropsychiatrySahlgrenska University HospitalRegion Västra GötalandGothenburgSweden
| | - Ariane Bollack
- The Grove CentreWhite Lion Road BuckinghamshireGE HealthCareAmershamUK
- Department of Medical Physics and BioengineeringCentre for Medical Image Computing (CMIC)University College LondonLondonUK
| | | | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Laboratory MedicineAmsterdam NeuroscienceNeurodegenerationAmsterdam UMC Vrije UniversiteitAmsterdamThe Netherlands
| | - Renaud La Joie
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Gil D. Rabinovici
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Nicolai Franzmeier
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Institute for Stroke and Dementia Research (ISD)University HospitalLMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Keith Johnson
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Center for Alzheimer Research and TreatmentBrigham and Women's HospitalBostonMassachusettsUSA
| | - Frederik Barkhof
- Department of Radiology and Nuclear MedicineVrije Universiteit AmsterdamAmsterdam University Medical CenterAmsterdamThe Netherlands
- Amsterdam NeuroscienceBrain imagingAmsterdamThe Netherlands
- UCL Queen Square Institute of Neurology and Center for Medical Image ComputingUniversity College LondonLondonUK
| | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Suzanne E. Schindler
- Department of NeurologyKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Lawrence S. Honig
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Alexis Moscoso Rial
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Nuclear Medicine Department and Molecular Imaging GroupInstituto de Investigación Sanitaria de Santiago de CompostelaSantiago de CompostelaSpain
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Department of NeuropsychiatrySahlgrenska University HospitalRegion Västra GötalandGothenburgSweden
- Dementia Research CentreInstitute of NeurologyUniversity College LondonLondonUK
| | - Henrik Zetterberg
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research InstituteUniversity College LondonLondonUK
- Hong Kong Center for Neurodegenerative DiseasesScience ParkHong KongChina
- Wisconsin Alzheimer's Disease Research CenterSchool of Medicine and Public HealthUniversity of WisconsinUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kaj Blennow
- The Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain InstituteICMPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiChina
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Gill Farrar
- The Grove CentreWhite Lion Road BuckinghamshireGE HealthCareAmershamUK
| |
Collapse
|
4
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated Appropriate Use Criteria for Amyloid and Tau PET: A Report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. J Nucl Med 2025:jnumed.124.268756. [PMID: 39778970 DOI: 10.2967/jnumed.124.268756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 01/11/2025] Open
Abstract
The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. Methods: The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. Results: For amyloid PET, 7 scenarios were rated as appropriate, 2 as uncertain, and 8 as rarely appropriate. For tau PET, 5 scenarios were rated as appropriate, 6 as uncertain, and 6 as rarely appropriate. Conclusion: AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Gil D Rabinovici
- Department of Neurology and Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California;
| | - David S Knopman
- Mayo Clinic Neurology and Neurosurgery, Rochester, Minnesota
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra Clinic, Pamplona, Spain
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri; Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Kevin J Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Peter Herscovitch
- Positron Emission Tomography Department, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Phillip H Kuo
- Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Jennifer H Lingler
- Department of Health and Community Systems, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | | | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Stephen P Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine, Brown University, Providence, Rhode Island
- Butler Hospital Memory and Aging Program, Providence, Rhode Island
| | | | | | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Molecular Neuroimaging, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
5
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated appropriate use criteria for amyloid and tau PET: A report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. Alzheimers Dement 2025; 21:e14338. [PMID: 39776249 PMCID: PMC11772739 DOI: 10.1002/alz.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. METHODS The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. RESULTS For amyloid PET, seven scenarios were rated as appropriate, two as uncertain, and eight as rarely appropriate. For tau PET, five scenarios were rated as appropriate, six as uncertain, and six as rarely appropriate. DISCUSSION AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease. HIGHLIGHTS A multidisciplinary workgroup convened by the Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging updated the appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. The goal of these updated AUC is to assist clinicians in identifying clinical scenarios in which amyloid or tau PET may be useful for guiding the diagnosis and management of patients who have, or are at risk for, cognitive decline These updated AUC are intended for dementia specialists who spend a significant proportion of their clinical effort caring for patients with cognitive complaints, as well as serve as a general reference for a broader audience interested in implementation of amyloid and tau PET in clinical practice.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Department of Neurology and Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Javier Arbizu
- Department of Nuclear MedicineUniversity of Navarra ClinicPamplonaSpain
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer's Disease Research CenterWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Kevin J. Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Oskar Hansson
- Department of Clinical Sciences MalmöClinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory Clinic, Skåne University HospitalSkånes universitetssjukhusMalmöSweden
| | - Peter Herscovitch
- Positron Emission Tomography DepartmentNational Institutes of Health Clinical CenterBethesdaMarylandUSA
| | - Phillip H. Kuo
- Medical Imaging, Medicine, and Biomedical EngineeringUniversity of ArizonaTucsonArizonaUSA
| | - Jennifer H. Lingler
- Department of Health and Community SystemsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Satoshi Minoshima
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUtahUSA
| | | | - Julie C. Price
- Department of RadiologyMassachusetts General Hospital, BostonCharlestownMassachusettsUSA
| | - Stephen P. Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine at Brown UniversityProvidenceRhode IslandUSA
- Butler Hospital Memory and Aging ProgramProvidenceRhode IslandUSA
| | | | - Maria C. Carrillo
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Keith A. Johnson
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Molecular Neuroimaging, Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Departments of Neurology and RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
6
|
Hunter TR, Santos LE, Tovar-Moll F, De Felice FG. Alzheimer's disease biomarkers and their current use in clinical research and practice. Mol Psychiatry 2025; 30:272-284. [PMID: 39232196 DOI: 10.1038/s41380-024-02709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
While blood-based tests are readily available for various conditions, including cardiovascular diseases, type 2 diabetes, and common cancers, Alzheimer's disease (AD) and other neurodegenerative diseases lack an early blood-based screening test that can be used in primary care. Major efforts have been made towards the investigation of approaches that may lead to minimally invasive, cost-effective, and reliable tests capable of measuring brain pathological status. Here, we review past and current technologies developed to investigate biomarkers of AD, including novel blood-based approaches and the more established cerebrospinal fluid and neuroimaging biomarkers of disease. The utility of blood as a source of AD-related biomarkers in both clinical practice and interventional trials is discussed, supported by a comprehensive list of clinical trials for AD drugs and interventions that list biomarkers as primary or secondary endpoints. We highlight that identifying individuals in early preclinical AD using blood-based biomarkers will improve clinical trials and the optimization of therapeutic treatments as they become available. Lastly, we discuss challenges that remain in the field and address new approaches being developed, such as the examination of cargo packaged within extracellular vesicles of neuronal origin isolated from peripheral blood.
Collapse
Affiliation(s)
- Tai R Hunter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Luis E Santos
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
| | | | - Fernanda G De Felice
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil.
- Centre for Neuroscience Studies and Department of Psychiatry, Queen's University, Kingston, ON, Canada.
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
7
|
Simrén J, Ashton NJ, Suárez-Calvet M, Zetterberg H. Alzheimer's disease-Biomarkers, clinical evaluation or both? J Neuropsychol 2024. [PMID: 39543822 DOI: 10.1111/jnp.12401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Affiliation(s)
- Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, UCL, London, UK
- UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
8
|
Puig-Pijoan A, García-Escobar G, Fernández-Lebrero A, Manero-Borràs RM, Sánchez-Benavides G, Navalpotro-Gómez I, Cascales Lahoz D, Suárez-Calvet M, Grau-Rivera O, Boltes Alandí A, Pont-Sunyer MC, Ortiz-Gil J, Carrillo-Molina S, López-Villegas D, Abellán-Vidal MT, Martínez-Casamitjana MI, Hernández-Sánchez JJ, Peña-Casanova J, Roquer J, Padrós Fluvià A, Puente-Périz V. The CORCOBIA study: Cut-off points of Alzheimer's disease CSF biomarkers in a clinical cohort. Neurologia 2024; 39:756-765. [PMID: 35961506 DOI: 10.1016/j.nrleng.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/24/2022] [Indexed: 10/15/2022] Open
Abstract
INTRODUCTION The analysis of the core biomarkers of Alzheimer's Disease (AD) in the cerebrospinal fluid (CSF) is recommended in the clinical units where it is available. Because of the absence of universal validated values, the determination of specific cut-off points for each center and its population is recommended. The main objective of the CORCOBIA study was to determine the cut-off points of core AD CSF biomarkers for several centers (Parc de Salut Mar, Barcelona and Hospital General de Granollers), which work with the same reference laboratory (Laboratori de Referència de Catalunya). METHODS Prospective study including cognitively unimpaired individuals (CU, n = 42), subjects with amnestic mild cognitive impairment (aMCI, n = 35) and patients with dementia due to Alzheimer's Disease (AD, n = 48), in whom clinical and neuropsychological assessment, neuroimaging, APOE genotyping and lumbar puncture to analyse amyloid beta peptides (Aβ42, Aβ40), total tau (tTau) and phosphorylated Tau (pTau181) using the Lumipulse G600II (Fujirebio) was performed. The values of sensitivity (SE), specificity (SP), predictive values and area under the curve (AUC) were calculated, determining the cut-off point according to the Youden index by comparing the CU and AD groups. RESULTS The resulting cut-offs and their AUC were the following: Aβ42 750 pg/mL (AUC 0.809); Aβ42/Aβ40 0.062 (AUC 0.78); pTau181 69.85 pg/mL (AUC 0.81); tTau 522.0 pg/mL (AUC 0.79); Aβ42/tTau 1.76 (AUC 0.86); Aβ42/pTau181 10.25 (AUC 0.86). CONCLUSIONS The determination of cut-off points of core AD CSF biomarkers for the participating centers allows a better diagnostic accuracy. The ratio CSF Aβ42/pTau181 shows the highest AUC and better balance between sensitivity and specificity.
Collapse
Affiliation(s)
- A Puig-Pijoan
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Farmacologia Integrada i Neurociència de Sistemes, Programa de Neurociències, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.
| | - G García-Escobar
- Farmacologia Integrada i Neurociència de Sistemes, Programa de Neurociències, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - A Fernández-Lebrero
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Farmacologia Integrada i Neurociència de Sistemes, Programa de Neurociències, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain; Barcelonaβeta Brain Research Center (BBRC), Fundació Pasqual Maragall, Barcelona, Spain
| | - R M Manero-Borràs
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Farmacologia Integrada i Neurociència de Sistemes, Programa de Neurociències, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - G Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Fundació Pasqual Maragall, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Hospital del Mar, Barcelona, Spain
| | - I Navalpotro-Gómez
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Farmacologia Integrada i Neurociència de Sistemes, Programa de Neurociències, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain; Barcelonaβeta Brain Research Center (BBRC), Fundació Pasqual Maragall, Barcelona, Spain
| | - D Cascales Lahoz
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - M Suárez-Calvet
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Barcelonaβeta Brain Research Center (BBRC), Fundació Pasqual Maragall, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Hospital del Mar, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - O Grau-Rivera
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Barcelonaβeta Brain Research Center (BBRC), Fundació Pasqual Maragall, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Hospital del Mar, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - A Boltes Alandí
- Servei de Neurologia, Hospital General de Granollers, Granollers, Barcelona, Spain
| | - M C Pont-Sunyer
- Servei de Neurologia, Hospital General de Granollers, Granollers, Barcelona, Spain
| | - J Ortiz-Gil
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain; Unitat de Psicologia, Hospital General de Granollers, Granollers, Barcelona, Spain; Fundación para la Investigación y Docencia Maria Angustias Gimenez (FIDMAG), Sant Boi de Llobregat, Barcelona, Spain
| | - S Carrillo-Molina
- Servei de Neurologia, Hospital General de Granollers, Granollers, Barcelona, Spain; Unitat de Psicologia, Hospital General de Granollers, Granollers, Barcelona, Spain
| | - D López-Villegas
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Hospital del Mar, Barcelona, Spain; Centre Emili Mira, Institut de Neuropsiquiatria i Addiccions (INAD), Parc de Salut Mar, Santa Coloma de Gramenet, Barcelona, Spain
| | - M T Abellán-Vidal
- Centre Emili Mira, Institut de Neuropsiquiatria i Addiccions (INAD), Parc de Salut Mar, Santa Coloma de Gramenet, Barcelona, Spain
| | - M I Martínez-Casamitjana
- Centre Emili Mira, Institut de Neuropsiquiatria i Addiccions (INAD), Parc de Salut Mar, Santa Coloma de Gramenet, Barcelona, Spain
| | | | - J Peña-Casanova
- Farmacologia Integrada i Neurociència de Sistemes, Programa de Neurociències, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - J Roquer
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Hospital del Mar, Barcelona, Spain
| | - A Padrós Fluvià
- Laboratori de Referència de Catalunya, Sant Boi de Llobregat, Barcelona, Spain
| | - V Puente-Périz
- Unitat de Deteriorament Cognitiu i Transtorns del Moviment, Servei de Neurologia, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain; Farmacologia Integrada i Neurociència de Sistemes, Programa de Neurociències, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.
| |
Collapse
|
9
|
Sandoval C, Lee J, Toth B, Nagaraj R, Schauer SP, Hoffman J, Calderon E, Kollmorgen G, Sanabria Bohórquez SM, Monteiro C, Teng E, Hanson JE, Yeh FL, Gutierrez J, Biever A. CSF complement proteins are elevated in prodromal to moderate Alzheimer's disease patients and are not altered by the anti-tau antibody semorinemab. Alzheimers Dement 2024; 20:7940-7953. [PMID: 39369294 PMCID: PMC11567840 DOI: 10.1002/alz.14271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/14/2024] [Accepted: 08/27/2024] [Indexed: 10/07/2024]
Abstract
INTRODUCTION Growing evidence suggests a role for neuroinflammation in Alzheimer's disease (AD). We investigated complement pathway activity in AD patient cerebrospinal fluid (CSF) and evaluated its modulation by the anti-tau antibody semorinemab. METHODS Immunoassays were applied to measure CSF complement proteins C4, factor B (FB), C3 and their cleavage fragments C4a, C3a, and factor Bb (Bb) in AD patients and a separate cognitively unimpaired (CU) cohort. RESULTS All measured CSF complement proteins were increased in AD versus CU subjects, with C4a displaying the most robust increase. Finally, semorinemab did not have a significant pharmacodynamic effect on CSF complement proteins. DISCUSSION Elevated levels of CSF C4a, C4, C3a, C3, Bb, and FB are consistent with complement activation in AD brains. Despite showing a reduction in CSF soluble tau species, semorinemab did not impact complement protein levels or activity. Further studies are needed to determine the value of complement proteins as neuroinflammation biomarkers in AD. HIGHLIGHTS Cerebrospinal fluid (CSF) complement proteins C4a, C3a, Bb, C4, C3, and factor B levels were increased in Alzheimer's disease (AD) patients compared to a separate cognitively unimpaired (CU) cohort. Baseline CSF complement protein levels were correlated with neuro-axonal degeneration and glial activation biomarkers in AD patients. The investigational anti-tau antibody semorinemab did not impact CSF complement protein levels or activity relative to the placebo arm.
Collapse
Affiliation(s)
- Cosme Sandoval
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Julie Lee
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Balazs Toth
- Department of Data and Statistical SciencesGenentech IncImmunology, Infectious Diseases and OpthalmologySouth San FranciscoCaliforniaUSA
| | - Rajini Nagaraj
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Stephen P. Schauer
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Jennifer Hoffman
- Department of Translational MedicineGenentech IncOperations Diagnostics Technology InnovationSouth San FranciscoCaliforniaUSA
| | - Emilia Calderon
- Department of Translational MedicineGenentech IncOperations Diagnostics Technology InnovationSouth San FranciscoCaliforniaUSA
| | | | | | - Cecilia Monteiro
- Department of Early Clinical DevelopmentGenentech IncSouth San FranciscoCaliforniaUSA
| | - Edmond Teng
- Department of Early Clinical DevelopmentGenentech IncSouth San FranciscoCaliforniaUSA
| | - Jesse E. Hanson
- Department of NeuroscienceGenentech IncSouth San FranciscoCaliforniaUSA
| | - Felix L. Yeh
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Johnny Gutierrez
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| | - Anne Biever
- Department of Translational MedicineGenentech IncOMNISouth San FranciscoCaliforniaUSA
| |
Collapse
|
10
|
Lowe VJ, Mester CT, Lundt ES, Lee J, Ghatamaneni S, Algeciras‐Schimnich A, Campbell MR, Graff‐Radford J, Nguyen A, Min H, Senjem ML, Machulda MM, Schwarz CG, Dickson DW, Murray ME, Kandimalla KK, Kantarci K, Boeve B, Vemuri P, Jones DT, Knopman D, Jack CR, Petersen RC, Mielke MM. Amyloid PET detects the deposition of brain Aβ earlier than CSF fluid biomarkers. Alzheimers Dement 2024; 20:8097-8112. [PMID: 39392211 PMCID: PMC12060152 DOI: 10.1002/alz.14317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Understanding the relationship between amyloid beta (Aβ) positron emission tomography (PET) and Aβ cerebrospinal fluid (CSF) biomarkers will define their potential utility in Aβ treatment. Few population-based or neuropathologic comparisons have been reported. METHODS Participants 50+ years with Aβ PET and Aβ CSF biomarkers (phosphorylated tau [p-tau]181/Aβ42, n = 505, and Aβ42/40, n = 54) were included from the Mayo Clinic Study on Aging. From these participants, an autopsy subgroup was identified (n = 47). The relationships of Aβ PET and Aβ CSF biomarkers were assessed cross-sectionally in all participants and longitudinally in autopsy data. RESULTS Cross-sectionally, more participants were Aβ PET+ versus Aβ CSF- than Aβ PET- versus Aβ CSF+ with an incremental effect when using Aβ PET regions selected for early Aβ deposition. The sensitivity for the first detection of Thal phase ≥ 1 in longitudinal data was higher for Aβ PET (89%) than p-tau181/Aβ42 (64%). DISCUSSION Aβ PET can detect earlier cortical Aβ deposition than Aβ CSF biomarkers. Aβ PET+ versus Aβ CSF- findings are several-fold greater using regional Aβ PET analyses and in peri-threshold-standardized uptake value ratio participants. HIGHLIGHTS Amyloid beta (Aβ) positron emission tomography (PET) has greater sensitivity for Aβ deposition than Aβ cerebrospinal fluid (CSF) in early Aβ development. A population-based sample of participants (n = 505) with PET and CSF tests was used. Cortical regions showing early Aβ on Aβ PET were also used in these analyses. Neuropathology was used to validate detection of Aβ by Aβ PET and Aβ CSF biomarkers.
Collapse
Affiliation(s)
- Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | - Carly T. Mester
- Departments of Radiology and Health Sciences ResearchMayo ClinicRochesterMinnesotaUSA
| | - Emily S. Lundt
- Departments of Radiology and Health Sciences ResearchMayo ClinicRochesterMinnesotaUSA
| | - Jeyeon Lee
- Department of Biomedical EngineeringCollege of MedicineHanyang UniversitySeoulRepublic of Korea
| | | | | | | | | | - Aivi Nguyen
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Hoon‐Ki Min
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | - Matthew L. Senjem
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
- Department of Information TechnologyMayo ClinicRochesterMinnesotaUSA
| | | | | | | | | | - Karunya K. Kandimalla
- Department of Pharmaceutics and Brain Barriers Research CenterUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Bradley Boeve
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | | | | | - David Knopman
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | | | | | - Michelle M. Mielke
- Department of Epidemiology and Prevention at Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
11
|
Pyun JM, Park YH, Kang MJ, Kim S. Cholinesterase inhibitor use in amyloid PET-negative mild cognitive impairment and cognitive changes. Alzheimers Res Ther 2024; 16:210. [PMID: 39358798 PMCID: PMC11448210 DOI: 10.1186/s13195-024-01580-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Cholinesterase inhibitors (ChEIs) are prescribed for Alzheimer's disease (AD) and sometimes for mild cognitive impairment (MCI) without knowing underlying pathologies and its effect on cognition. We investigated the frequency of ChEI prescriptions in amyloid-negative MCI and their association with cognitive changes in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. METHODS We included participants with amyloid positron emission tomography (PET)-negative MCI from the ADNI. We analyzed the associations of ChEI use with cognitive changes, brain volume, and cerebrospinal fluid (CSF) total tau (t-tau), hyperphosphorylated tau181 (p-tau181), and p-tau181/t-tau ratio. RESULTS ChEIs were prescribed in 27.4% of amyloid PET-negative MCI and were associated with faster cognitive decline, reduced baseline hippocampal volume and entorhinal cortical thickness, and a longitudinal decrease in the frontal lobe cortical thickness. CONCLUSIONS The association between ChEI use and accelerated cognitive decline may stem from underlying pathologies involving reduced hippocampal volume, entorhinal cortical thickness and faster frontal lobe atrophy. We suggest that ChEI use in amyloid PET-negative MCI patients might need further consideration, and studies investigating the causality between ChEI use and cognitive decline are warranted in the future.
Collapse
Affiliation(s)
- Jung-Min Pyun
- Department of Neurology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, 59, Daesagwan-ro, Yongsan-gu, Seoul, 04401, Republic of Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Republic of Korea
| | - Min Ju Kang
- Department of Neurology, Veterans Health Service Medical Center, 53, Jinhwangdo-ro 61-gil, Gangdong-gu, Seoul, 05368, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, 13620, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
12
|
Leuzy A, Raket LL, Villemagne VL, Klein G, Tonietto M, Olafson E, Baker S, Saad ZS, Bullich S, Lopresti B, Bohorquez SS, Boada M, Betthauser TJ, Charil A, Collins EC, Collins JA, Cullen N, Gunn RN, Higuchi M, Hostetler E, Hutchison RM, Iaccarino L, Insel PS, Irizarry MC, Jack CR, Jagust WJ, Johnson KA, Johnson SC, Karten Y, Marquié M, Mathotaarachchi S, Mintun MA, Ossenkoppele R, Pappas I, Petersen RC, Rabinovici GD, Rosa‐Neto P, Schwarz CG, Smith R, Stephens AW, Whittington A, Carrillo MC, Pontecorvo MJ, Haeberlein SB, Dunn B, Kolb HC, Sivakumaran S, Rowe CC, Hansson O, Doré V. Harmonizing tau positron emission tomography in Alzheimer's disease: The CenTauR scale and the joint propagation model. Alzheimers Dement 2024; 20:5833-5848. [PMID: 39041435 PMCID: PMC11497758 DOI: 10.1002/alz.13908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Tau-positron emission tomography (PET) outcome data of patients with Alzheimer's disease (AD) cannot currently be meaningfully compared or combined when different tracers are used due to differences in tracer properties, instrumentation, and methods of analysis. METHODS Using head-to-head data from five cohorts with tau PET radiotracers designed to target tau deposition in AD, we tested a joint propagation model (JPM) to harmonize quantification (units termed "CenTauR" [CTR]). JPM is a statistical model that simultaneously models the relationships between head-to-head and anchor point data. JPM was compared to a linear regression approach analogous to the one used in the amyloid PET Centiloid scale. RESULTS A strong linear relationship was observed between CTR values across brain regions. Using the JPM approach, CTR estimates were similar to, but more accurate than, those derived using the linear regression approach. DISCUSSION Preliminary findings using the JPM support the development and adoption of a universal scale for tau-PET quantification. HIGHLIGHTS Tested a novel joint propagation model (JPM) to harmonize quantification of tau PET. Units of common scale are termed "CenTauRs". Tested a Centiloid-like linear regression approach. Using five cohorts with head-to-head tau PET, JPM outperformed linearregressionbased approach. Strong linear relationship was observed between CenTauRs values across brain regions.
Collapse
|
13
|
Arnold SE, Hendrix S, Nicodemus‐Johnson J, Knowlton N, Williams VJ, Burns JM, Crane M, McManus AJ, Vaishnavi SN, Arvanitakis Z, Neugroschl J, Bell K, Trombetta BA, Carlyle BC, Kivisäkk P, Dodge HH, Tanzi RE, Yeramian PD, Leslie K. Biological effects of sodium phenylbutyrate and taurursodiol in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12487. [PMID: 39131742 PMCID: PMC11310855 DOI: 10.1002/trc2.12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/25/2024] [Accepted: 05/13/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Sodium phenylbutyrate and taurursodiol (PB and TURSO) is hypothesized to mitigate endoplasmic reticulum stress and mitochondrial dysfunction, two of many mechanisms implicated in Alzheimer's disease (AD) pathophysiology. METHODS The first-in-indication phase 2a PEGASUS trial was designed to gain insight into PB and TURSO effects on mechanistic targets of engagement and disease biology in AD. The primary clinical efficacy outcome was a global statistical test combining three endpoints relevant to disease trajectory (cognition [Mild/Moderate Alzheimer's Disease Composite Score], function [Functional Activities Questionnaire], and total hippocampal volume on magnetic resonance imaging). Secondary clinical outcomes included various cognitive, functional, and neuropsychiatric assessments. Cerebrospinal fluid (CSF) biomarkers spanning multiple pathophysiological pathways in AD were evaluated in participants with both baseline and Week 24 samples (exploratory outcome). RESULTS PEGASUS enrolled 95 participants (intent-to-treat [ITT] cohort); cognitive assessments indicated significantly greater baseline cognitive impairment in the PB and TURSO (n = 51) versus placebo (n = 44) group. Clinical efficacy outcomes did not significantly differ between treatment groups in the ITT cohort. CSF interleukin-15 increased from baseline to Week 24 within the placebo group (n = 34). In the PB and TURSO group (n = 33), reductions were observed in core AD biomarkers phosphorylated tau-181 (p-tau181) and total tau; synaptic and neuronal degeneration biomarkers neurogranin and fatty acid binding protein-3 (FABP3); and gliosis biomarker chitinase 3-like protein 1 (YKL-40), while the oxidative stress marker 8-hydroxy-2-deoxyguanosine (8-OHdG) increased. Between-group differences were observed for the Aβ42/40 ratio, p-tau181, total tau, neurogranin, FABP3, YKL-40, interleukin-15, and 8-OHdG. Additional neurodegeneration, inflammation, and metabolic biomarkers showed no differences between groups. DISCUSSION While between-group differences in clinical outcomes were not observed, most likely due to the small sample size and relatively short treatment duration, exploratory biomarker analyses suggested that PB and TURSO engages multiple pathophysiologic pathways in AD. Highlights Proteostasis and mitochondrial stress play key roles in Alzheimer's disease (AD).Sodium phenylbutyrate and taurursodiol (PB and TURSO) targets these mechanisms.The PEGASUS trial was designed to assess PB and TURSO effects on biologic AD targets.PB and TURSO reduced exploratory biomarkers of AD and neurodegeneration.Supports further clinical development of PB and TURSO in neurodegenerative diseases.
Collapse
Affiliation(s)
- Steven E. Arnold
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | | | | | - Victoria J. Williams
- Department of MedicineUniversity of Wisconsin‐MadisonSchool of Medicine and Public HealthMadisonWisconsinUSA
| | - Jeffrey M. Burns
- University of Kansas Alzheimer's Disease Research CenterFairwayKansasUSA
| | - Monica Crane
- Genesis Neuroscience ClinicKnoxvilleTennesseeUSA
| | - Alison J. McManus
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Sanjeev N. Vaishnavi
- Department of NeurologyPenn Memory CenterPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Judith Neugroschl
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Karen Bell
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Bianca A. Trombetta
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Becky C. Carlyle
- Department of PhysiologyAnatomy & Genetics and Kavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Pia Kivisäkk
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Hiroko H. Dodge
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Rudolph E. Tanzi
- Department of NeurologyGenetics and Aging Research UnitMcCance Center for Brain HealthMassachusetts General HospitalHarvard UniversityBostonMassachusettsUSA
| | | | - Kent Leslie
- Amylyx Pharmaceuticals, Inc.CambridgeMassachusettsUSA
- Present address:
Division of Biology and Biological Engineering Graduate ProgramCalifornia Institute of TechnologyPasadenaCAUSA
| |
Collapse
|
14
|
Yu Y, Yu S, Battaglia G, Tian X. Amyloid-β in Alzheimer's disease: Structure, toxicity, distribution, treatment, and prospects. IBRAIN 2024; 10:266-289. [PMID: 39346788 PMCID: PMC11427815 DOI: 10.1002/ibra.12155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 10/01/2024]
Abstract
Amyloid-β (Aβ) is a pivotal biomarker in Alzheimer's disease (AD), attracting considerable attention from numerous researchers. There is uncertainty regarding whether clearing Aβ is beneficial or harmful to cognitive function. This question has been a central topic of research, especially given the lack of success in developing Aβ-targeted drugs for AD. However, with the Food and Drug Administration's approval of Lecanemab as the first anti-Aβ medication in July 2023, there is a significant shift in perspective on the potential of Aβ as a therapeutic target for AD. In light of this advancement, this review aims to illustrate and consolidate the molecular structural attributes and pathological ramifications of Aβ. Furthermore, it elucidates the determinants influencing its expression levels while delineating the gamut of extant Aβ-targeted pharmacotherapies that have been subjected to clinical or preclinical evaluation. Subsequently, a comprehensive analysis is presented, dissecting the research landscape of Aβ across the domains above, culminating in the presentation of informed perspectives. Concluding reflections contemplate the supplementary advantages conferred by nanoparticle constructs, conceptualized within the framework of multivalent theory, within the milieu of AD diagnosis and therapeutic intervention, supplementing conventional modalities.
Collapse
Affiliation(s)
- Yifan Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Shilong Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Giuseppe Battaglia
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Xiaohe Tian
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
15
|
Dakterzada F, Cipriani R, López-Ortega R, Arias A, Riba-Llena I, Ruiz-Julián M, Huerto R, Tahan N, Matute C, Capetillo-Zarate E, Piñol-Ripoll G. Assessment of the Correlation and Diagnostic Accuracy between Cerebrospinal Fluid and Plasma Alzheimer's Disease Biomarkers: A Comparison of the Lumipulse and Simoa Platforms. Int J Mol Sci 2024; 25:4594. [PMID: 38731812 PMCID: PMC11083365 DOI: 10.3390/ijms25094594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
We compared the clinical and analytical performance of Alzheimer's disease (AD) plasma biomarkers measured using the single-molecule array (Simoa) and Lumipulse platforms. We quantified the plasma levels of amyloid beta 42 (Aβ42), Aβ40, phosphorylated tau (Ptau181), and total tau biomarkers in 81 patients with mild cognitive impairment (MCI), 30 with AD, and 16 with non-AD dementia. We found a strong correlation between the Simoa and Lumipulse methods. Concerning the clinical diagnosis, Simoa Ptau181/Aβ42 (AUC 0.739, 95% CI 0.592-0.887) and Lumipulse Aβ42 and Ptau181/Aβ42 (AUC 0.735, 95% CI 0.589-0.882 and AUC 0.733, 95% CI 0.567-0.900) had the highest discriminating power. However, their power was significantly lower than that of CSF Aβ42/Aβ40, as measured by Lumipulse (AUC 0.879, 95% CI 0.766-0.992). Simoa Ptau181 and Lumipulse Ptau181/Aβ42 were the markers most consistent with the CSF Aβ42/Aβ40 status (AUC 0.801, 95% CI 0.712-0.890 vs. AUC 0.870, 95% CI 0.806-0.934, respectively) at the ≥2.127 and ≥0.084 cut-offs, respectively. The performance of the Simoa and Lumipulse plasma AD assays is weaker than that of CSF AD biomarkers. At present, the analysed AD plasma biomarkers may be useful for screening to reduce the number of lumbar punctures in the clinical setting.
Collapse
Affiliation(s)
- Farida Dakterzada
- Cognitive Disorders Unit, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, 25198 Lleida, Spain; (F.D.); (A.A.); (I.R.-L.); (M.R.-J.); (R.H.); (N.T.)
| | - Raffaela Cipriani
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; (R.C.); (C.M.); (E.C.-Z.)
| | - Ricard López-Ortega
- Laboratori ClínicInstitut Català de la Salut (ICS), Hospital Universitari Arnau de Vilanova, 25198 Lleida, Spain;
| | - Alfonso Arias
- Cognitive Disorders Unit, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, 25198 Lleida, Spain; (F.D.); (A.A.); (I.R.-L.); (M.R.-J.); (R.H.); (N.T.)
| | - Iolanda Riba-Llena
- Cognitive Disorders Unit, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, 25198 Lleida, Spain; (F.D.); (A.A.); (I.R.-L.); (M.R.-J.); (R.H.); (N.T.)
| | - Maria Ruiz-Julián
- Cognitive Disorders Unit, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, 25198 Lleida, Spain; (F.D.); (A.A.); (I.R.-L.); (M.R.-J.); (R.H.); (N.T.)
| | - Raquel Huerto
- Cognitive Disorders Unit, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, 25198 Lleida, Spain; (F.D.); (A.A.); (I.R.-L.); (M.R.-J.); (R.H.); (N.T.)
| | - Nuria Tahan
- Cognitive Disorders Unit, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, 25198 Lleida, Spain; (F.D.); (A.A.); (I.R.-L.); (M.R.-J.); (R.H.); (N.T.)
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; (R.C.); (C.M.); (E.C.-Z.)
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- CIBERNED, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, 28029 Madrid, Spain
| | - Estibaliz Capetillo-Zarate
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; (R.C.); (C.M.); (E.C.-Z.)
- CIBERNED, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas, 28029 Madrid, Spain
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01008 Vitoria-Gasteiz, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Gerard Piñol-Ripoll
- Cognitive Disorders Unit, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, 25198 Lleida, Spain; (F.D.); (A.A.); (I.R.-L.); (M.R.-J.); (R.H.); (N.T.)
- Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida (UDL), 25002 Lleida, Spain
| |
Collapse
|
16
|
Pyun JM, Park YH, Youn YC, Kang MJ, Shim KH, Jang JW, You J, Nho K, Kim S. Characteristics of discordance between amyloid positron emission tomography and plasma amyloid-β 42/40 positivity. Transl Psychiatry 2024; 14:88. [PMID: 38341444 PMCID: PMC10858862 DOI: 10.1038/s41398-024-02766-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/12/2024] Open
Abstract
Various plasma biomarkers for amyloid-β (Aβ) have shown high predictability of amyloid PET positivity. However, the characteristics of discordance between amyloid PET and plasma Aβ42/40 positivity are poorly understood. Thorough interpretation of discordant cases is vital as Aβ plasma biomarker is imminent to integrate into clinical guidelines. We aimed to determine the characteristics of discordant groups between amyloid PET and plasma Aβ42/40 positivity, and inter-assays variability depending on plasma assays. We compared tau burden measured by PET, brain volume assessed by MRI, cross-sectional cognitive function, longitudinal cognitive decline and polygenic risk score (PRS) between PET/plasma groups (PET-/plasma-, PET-/plasma+, PET+/plasma-, PET+/plasma+) using Alzheimer's Disease Neuroimaging Initiative database. Additionally, we investigated inter-assays variability between immunoprecipitation followed by mass spectrometry method developed at Washington University (IP-MS-WashU) and Elecsys immunoassay from Roche (IA-Elc). PET+/plasma+ was significantly associated with higher tau burden assessed by PET in entorhinal, Braak III/IV, and Braak V/VI regions, and with decreased volume of hippocampal and precuneus regions compared to PET-/plasma-. PET+/plasma+ showed poor performances in global cognition, memory, executive and daily-life function, and rapid cognitive decline. PET+/plasma+ was related to high PRS. The PET-/plasma+ showed intermediate changes between PET-/plasma- and PET+/plasma+ in terms of tau burden, hippocampal and precuneus volume, cross-sectional and longitudinal cognition, and PRS. PET+/plasma- represented heterogeneous characteristics with most prominent variability depending on plasma assays. Moreover, IP-MS-WashU showed more linear association between amyloid PET standardized uptake value ratio and plasma Aβ42/40 than IA-Elc. IA-Elc showed more plasma Aβ42/40 positivity in the amyloid PET-negative stage than IP-MS-WashU. Characteristics of PET-/plasma+ support plasma biomarkers as early biomarker of amyloidopathy prior to amyloid PET. Various plasma biomarker assays might be applied distinctively to detect different target subjects or disease stages.
Collapse
Affiliation(s)
- Jung-Min Pyun
- Department of Neurology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, 59, Daesagwan-ro, Yongsan-gu, Seoul, 04401, Republic of Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University Hospital, 102, Heukseok-ro, Dongjak-gu, Seoul, 06973, Republic of Korea
| | - Min Ju Kang
- Department of Neurology, Veterans Health Service Medical Center, 53, Jinhwangdo-ro 61-gil, Gangdong-gu, Seoul, 05368, Republic of Korea
| | - Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, 1342, Seongnamdaero, Sujeong-gu, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University Hospital, Kangwon National University College of Medicine, 156, Baengnyeong-ro, Chuncheon-si, Gangwon-do, 24289, Republic of Korea
| | - Jihwan You
- Department of Neurology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, 59, Daesagwan-ro, Yongsan-gu, Seoul, 04401, Republic of Korea
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, 355 W 16th, GH 4101, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 410 W 10th, Health Information and Translational Science Building, Suite 5000, Indianapolis, IN, 46202, USA
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.
| |
Collapse
|
17
|
Sánchez-Soblechero A, López-García S, Lage C, Fernández-Matarrubia M, Irure J, López-Hoyos M, Jiménez-Bonilla J, Quirce R, de Arcocha-Torres M, Cuenca-Vera O, Martín-Arroyo J, Martínez-Dubarbie F, Pozueta A, García-Martínez M, Infante J, Sánchez-Juan P, Rodríguez-Rodríguez E. Where Should I Draw the Line: PET-Driven, Data-Driven, or Manufacturer Cut-Off? J Alzheimers Dis 2024; 98:957-967. [PMID: 38489172 DOI: 10.3233/jad-230678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Background The optimal cut-off for Alzheimer's disease (AD) CSF biomarkers remains controversial. Objective To analyze the performance of cut-off points standardized by three methods: one that optimized the agreement between 11C-Pittsburgh compound B PET (a-PET) and CSF biomarkers (Aβ1-42, pTau, tTau, and Aβ1-42/Aβ1-40 ratio) in our population, called PET-driven; an unbiased cut-off using data from a healthy research cohort, called data-driven, and that provided by the manufacturer. We also compare changes in ATN classification. Methods CSF biomarkers measured by the LUMIPULSE G600II platform and qualitative visualization of amyloid positron emission tomography (a-PET) were performed in all the patients. We established a cut-off for each single biomarker and Aβ1-42/Aβ1-40 ratio that optimized their agreement with a-PET using ROC curves. Sensitivity, Specificity, and Overall Percent of Agreement are assessed using a-PET or clinical diagnosis as gold standard for every cut-off. Also, we established a data-driven cut-off from our cognitively unimpaired cohort. We then analyzed changes in ATN classification. Results One hundred and ten patients were recruited. Sixty-six (60%) were a-PET positive. PET-driven cut-offs were: pTau > 57, tTau > 362.62, Aβ1-42/Aβ1-40 < 0.069. For a single biomarker, pTau showed the highest accuracy (AUC 0.926). New PET-driven cut-offs classified patients similarly to manufacturer cut-offs (only two patients changed). However, 20 patients (18%) changed when data-driven cut-offs were used. Conclusions We established our sample's best CSF biomarkers cut-offs using a-PET as the gold standard. These cut-offs categorize better symptomatic subjects than data-driven in ATN classification, but they are very similar to the manufacturer's.
Collapse
Affiliation(s)
| | - Sara López-García
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carmen Lage
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marta Fernández-Matarrubia
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Irure
- Immunology Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| | - Julio Jiménez-Bonilla
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Remedios Quirce
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - María de Arcocha-Torres
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Oriana Cuenca-Vera
- Nuclear Medicine Department, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Juan Martín-Arroyo
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
| | - Francisco Martínez-Dubarbie
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ana Pozueta
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María García-Martínez
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jon Infante
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| | - Pascual Sánchez-Juan
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, Madrid, Spain
| | - Eloy Rodríguez-Rodríguez
- Neurology Department, Cognitive Impairment Unit, 'Marqués de Valdecilla' University Hospital, Santander, Spain
- Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
| |
Collapse
|
18
|
Kang JH, Korecka M, Lee EB, Cousins KAQ, Tropea TF, Chen-Plotkin AA, Irwin DJ, Wolk D, Brylska M, Wan Y, Shaw LM. Alzheimer Disease Biomarkers: Moving from CSF to Plasma for Reliable Detection of Amyloid and tau Pathology. Clin Chem 2023; 69:1247-1259. [PMID: 37725909 PMCID: PMC10895336 DOI: 10.1093/clinchem/hvad139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/07/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Development of validated biomarkers to detect early Alzheimer disease (AD) neuropathology is needed for therapeutic AD trials. Abnormal concentrations of "core" AD biomarkers, cerebrospinal fluid (CSF) amyloid beta1-42, total tau, and phosphorylated tau correlate well with neuroimaging biomarkers and autopsy findings. Nevertheless, given the limitations of established CSF and neuroimaging biomarkers, accelerated development of blood-based AD biomarkers is underway. CONTENT Here we describe the clinical significance of CSF and plasma AD biomarkers to detect disease pathology throughout the Alzheimer continuum and correlate with imaging biomarkers. Use of the AT(N) classification by CSF and imaging biomarkers provides a more objective biologically based diagnosis of AD than clinical diagnosis alone. Significant progress in measuring CSF AD biomarkers using extensively validated highly automated assay systems has facilitated their transition from research use only to approved in vitro diagnostics tests for clinical use. We summarize development of plasma AD biomarkers as screening tools for enrollment and monitoring participants in therapeutic trials and ultimately in clinical care. Finally, we discuss the challenges for AD biomarkers use in clinical trials and precision medicine, emphasizing the possible ethnocultural differences in the levels of AD biomarkers. SUMMARY CSF AD biomarker measurements using fully automated analytical platforms is possible. Building on this experience, validated blood-based biomarker tests are being implemented on highly automated immunoassay and mass spectrometry platforms. The progress made developing analytically and clinically validated plasma AD biomarkers within the AT(N) classification scheme can accelerate use of AD biomarkers in therapeutic trials and routine clinical practice.
Collapse
Affiliation(s)
- Ju Hee Kang
- Department of Pharmacology and Clinical Pharmacology, Research Center for Controlling Intercellular Communication, Inha University, Incheon, South Korea
| | - Magdalena Korecka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katheryn A Q Cousins
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alice A Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Magdalena Brylska
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yang Wan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
19
|
Petersen RC, Graf A, Brady C, De Santi S, Florian H, Landen J, Pontecorvo M, Randolph C, Sink KM, Carrillo MC, Weber CJ. Operationalizing selection criteria for clinical trials in Alzheimer's disease: Biomarker and clinical considerations. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12434. [PMID: 38023620 PMCID: PMC10655199 DOI: 10.1002/trc2.12434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
Alzheimer's disease (AD) staging criteria lack standardized, empirical description. Well-defined AD staging criteria are an important consideration in protocol design, influencing a more standardized inclusion/exclusion criteria and defining what constitutes meaningful differentiation among the stages. However, many trials are being designed on the basis of biomarker features and the two need to be coordinated. The Alzheimer's Association Research Roundtable (AARR) Spring 2021 meeting discussed the implementation of preclinical AD staging criteria, and provided recommendations for how they may best be incorporated into clinical trials research. Discussion also included what currently available tools for global clinical trials may best define populations in preclinical AD trials, and if are we able to differentiate preclinical from clinical stages of the disease. Well-defined AD staging criteria are key to improving early detection, diagnostics, clinical trial enrollment, and identifying statistically significant clinical changes, and researchers discussed how emerging blood biomarkers may help with more efficient screening in preclinical stages.
Collapse
Affiliation(s)
| | - Ana Graf
- Novartis Pharma AGBaselSwitzerland
| | - Chris Brady
- WCG Clinical Endpoint Solutions, PrincetonNew JerseyUSA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Motta C, Di Donna MG, Bonomi CG, Assogna M, Chiaravalloti A, Mercuri NB, Koch G, Martorana A. Different associations between amyloid-βeta 42, amyloid-βeta 40, and amyloid-βeta 42/40 with soluble phosphorylated-tau and disease burden in Alzheimer's disease: a cerebrospinal fluid and fluorodeoxyglucose-positron emission tomography study. Alzheimers Res Ther 2023; 15:144. [PMID: 37649105 PMCID: PMC10466826 DOI: 10.1186/s13195-023-01291-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Despite the high sensitivity of cerebrospinal fluid (CSF) amyloid beta (Aβ)42 to detect amyloid pathology, the Aβ42/Aβ40 ratio (amyR) better estimates amyloid load, with higher specificity for Alzheimer's disease (AD). However, whether Aβ42 and amyR have different meanings and whether Aβ40 represents more than an Aβ42-corrective factor remain to be clarified. Our study aimed to compare the ability of Aβ42 and amyR to detect AD pathology in terms of p-tau/Aβ42 ratio and brain glucose metabolic patterns using fluorodeoxyglucose-positron emission tomography (FDG-PET). METHODS CSF biomarkers were analyzed with EUROIMMUN ELISA. We included 163 patients showing pathological CSF Aβ42 and normal p-tau (A + T - = 98) or pathological p-tau levels (A + T + = 65) and 36 control subjects (A - T -). A + T - patients were further stratified into those with normal (CSFAβ42 + /amyR - = 46) and pathological amyR (CSFAβ42 + /amyR + = 52). We used two distinct cut-offs to determine pathological values of p-tau/Aβ42: (1) ≥ 0.086 and (2) ≥ 0.122. FDG-PET patterns were evaluated in a subsample of patients (n = 46) and compared to 24 controls. RESULTS CSF Aβ40 levels were the lowest in A - T - and in CSFAβ42 + /amyR - , higher in CSFAβ42 + /amyR + and highest in A + T + (F = 50.75; p < 0.001), resembling CSF levels of p-tau (F = 192; p < 0.001). We found a positive association between Aβ40 and p-tau in A - T - (β = 0.58; p < 0.001), CSFAβ42 + /amyR - (β = 0.47; p < 0.001), and CSFAβ42 + /amyR + patients (β = 0.48; p < 0.001) but not in A + T + . Investigating biomarker changes as a function of amyR, we observed a weak variation in CSF p-tau (+ 2 z-scores) and Aβ40 (+ 0.8 z-scores) in the normal amyR range, becoming steeper over the pathological threshold of amyR (p-tau: + 5 z-scores, Aβ40: + 4.5 z-score). CSFAβ42 + /amyR + patients showed a significantly higher probability of having pathological p-tau/Aβ42 than CSFAβ42 + /amyR - (cut-off ≥ 0.086: OR 23.3; cut-off ≥ 0.122: OR 8.8), which however still showed pathological values of p-tau/Aβ42 in some cases (cut-off ≥ 0.086: 35.7%; cut-off ≥ 0.122: 17.3%) unlike A - T - . Accordingly, we found reduced FDG metabolism in the temporoparietal regions of CSFAβ42 + /amyR - compared to controls, and further reduction in frontal areas in CSFAβ42 + /amyR + , like in A + T + . CONCLUSIONS Pathological p-tau/Aβ42 and FDG hypometabolism typical of AD can be found in patients with decreased CSF Aβ42 levels alone. AmyR positivity, associated with higher Aβ40 levels, is accompanied by higher CSF p-tau and widespread FDG hypometabolism.
Collapse
Affiliation(s)
- Caterina Motta
- UOSD Centro Demenze, University of Rome "Tor Vergata", Rome, Italy.
| | | | | | - Martina Assogna
- UOSD Centro Demenze, University of Rome "Tor Vergata", Rome, Italy
- Experimental Neuropsychophysiology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Istituto Neurologico Mediterraneo, Pozzilli, Italy
| | | | - Giacomo Koch
- Experimental Neuropsychophysiology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Human Physiology Unit, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
21
|
Palmqvist S, Rossi M, Hall S, Quadalti C, Mattsson-Carlgren N, Dellavalle S, Tideman P, Pereira JB, Nilsson MH, Mammana A, Janelidze S, Baiardi S, Stomrud E, Parchi P, Hansson O. Cognitive effects of Lewy body pathology in clinically unimpaired individuals. Nat Med 2023; 29:1971-1978. [PMID: 37464059 PMCID: PMC10427420 DOI: 10.1038/s41591-023-02450-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
α-Synuclein aggregates constitute the pathology of Lewy body (LB) disease. Little is known about the effects of LB pathology in preclinical (presymptomatic) individuals, either as isolated pathology or coexisting with Alzheimer's disease (AD) pathology (β-amyloid (Aβ) and tau). We examined the effects of LB pathology using a cerebrospinal fluid α-synuclein-seed amplification assay in 1,182 cognitively and neurologically unimpaired participants from the BioFINDER study: 8% were LB positive, 26% Aβ positive (13% of those were LB positive) and 16% tau positive. LB positivity occurred more often in the presence of Aβ positivity but not tau positivity. LB pathology had independently negative effects on cross-sectional and longitudinal global cognition and memory and on longitudinal attention/executive function. Tau had cognitive effects of a similar magnitude, but these were less pronounced for Aβ. Participants with both LB and AD (Aβ and tau) pathology exhibited faster cognitive decline than those with only LB or AD pathology. LB, but not AD, pathology was associated with reduced sense of smell. Only LB-positive participants progressed to clinical LB disease over 10 years. These results are important for individualized prognosis, recruitment and choice of outcome measures in preclinical LB disease trials, but also for the design of early AD trials because >10% of individuals with preclinical AD have coexisting LB pathology.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Sara Hall
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Corinne Quadalti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sofia Dellavalle
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Maria H Nilsson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Department of Health Sciences, Lund University, Lund, Sweden
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Simone Baiardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
22
|
Salvadó G, Larsson V, Cody KA, Cullen NC, Jonaitis EM, Stomrud E, Kollmorgen G, Wild N, Palmqvist S, Janelidze S, Mattsson-Carlgren N, Zetterberg H, Blennow K, Johnson SC, Ossenkoppele R, Hansson O. Optimal combinations of CSF biomarkers for predicting cognitive decline and clinical conversion in cognitively unimpaired participants and mild cognitive impairment patients: A multi-cohort study. Alzheimers Dement 2023; 19:2943-2955. [PMID: 36648169 PMCID: PMC10350470 DOI: 10.1002/alz.12907] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/30/2022] [Accepted: 11/15/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Our objective was determining the optimal combinations of cerebrospinal fluid (CSF) biomarkers for predicting disease progression in Alzheimer's disease (AD) and other neurodegenerative diseases. METHODS We included 1,983 participants from three different cohorts with longitudinal cognitive and clinical data, and baseline CSF levels of Aβ42, Aβ40, phosphorylated tau at threonine-181 (p-tau), neurofilament light (NfL), neurogranin, α-synuclein, soluble triggering receptor expressed on myeloid cells 2 (sTREM2), glial fibrillary acidic protein (GFAP), YKL-40, S100b, and interleukin 6 (IL-6) (Elecsys NeuroToolKit). RESULTS Change of modified Preclinical Alzheimer's Cognitive Composite (mPACC) in cognitively unimpaired (CU) was best predicted by p-tau/Aβ42 alone (R2 ≥ 0.31) or together with NfL (R2 = 0.25), while p-tau/Aβ42 (R2 ≥ 0.19) was sufficient to accurately predict change of the Mini-Mental State Examination (MMSE) in mild cognitive impairment (MCI) patients. P-tau/Aβ42 (AUC ≥ 0.87) and p-tau/Aβ42 together with NfL (AUC ≥ 0.75) were the best predictors of conversion to AD and all-cause dementia, respectively. DISCUSSION P-tau/Aβ42 is sufficient for predicting progression in AD, with very high accuracy. Adding NfL improves the prediction of all-cause dementia conversion and cognitive decline.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Victoria Larsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Karly A Cody
- Wisconsin Alzheimer’s Disease Research Center University of Wisconsin School of Medicine and Public Health Madison Wisconsin, Madison, Wisconsin, USA
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Erin M Jonaitis
- Wisconsin Alzheimer’s Disease Research Center University of Wisconsin School of Medicine and Public Health Madison Wisconsin, Madison, Wisconsin, USA
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | | | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Disease Research Center University of Wisconsin School of Medicine and Public Health Madison Wisconsin, Madison, Wisconsin, USA
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research, Education and Clinical Center at the William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
23
|
Palmqvist S, Stomrud E, Cullen N, Janelidze S, Manuilova E, Jethwa A, Bittner T, Eichenlaub U, Suridjan I, Kollmorgen G, Riepe M, von Arnim CA, Tumani H, Hager K, Heidenreich F, Mattsson-Carlgren N, Zetterberg H, Blennow K, Hansson O. An accurate fully automated panel of plasma biomarkers for Alzheimer's disease. Alzheimers Dement 2023; 19:1204-1215. [PMID: 35950735 PMCID: PMC9918613 DOI: 10.1002/alz.12751] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/27/2022] [Accepted: 06/10/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION There is a great need for fully automated plasma assays that can measure amyloid beta (Aβ) pathology and predict future Alzheimer's disease (AD) dementia. METHODS Two cohorts (n = 920) were examined: Panel A+ (n = 32 cognitively unimpaired [CU], n = 106 mild cognitive impairment [MCI], and n = 89 AD) and BioFINDER-1 (n = 461 CU, n = 232 MCI). Plasma Aβ42/Aβ40, phosphorylated tau (p-tau)181, two p-tau217 variants, ApoE4 protein, neurofilament light, and GFAP were measured using Elecsys prototype immunoassays. RESULTS The best biomarker for discriminating Aβ-positive versus Aβ-negative participants was Aβ42/Aβ40 (are under the curve [AUC] 0.83-0.87). Combining Aβ42/Aβ40, p-tau181, and ApoE4 improved the AUCs significantly (0.90 to 0.93; P< 0.01). Adding additional biomarkers had marginal effects (ΔAUC ≤0.01). In BioFINDER, p-tau181, p-tau217, and ApoE4 predicted AD dementia within 6 years in CU (AUC 0.88) and p-tau181, p-tau217, and Aβ42/Aβ40 in MCI (AUC 0.87). DISCUSSION The high accuracies for Aβ pathology and future AD dementia using fully automated instruments are promising for implementing plasma biomarkers in clinical trials and clinical routine.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Nicholas Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
| | | | | | | | | | | | | | - Matthias Riepe
- Division of Geriatric Psychiatry, Ulm University, Germany
| | - Christine A.F. von Arnim
- Division of Geriatrics, University Medical Center Göttingen, Georg-August-University, Goettingen, Germany
| | | | - Klaus Hager
- Institute for General Medicine and Palliative Medicine, Hannover Medical School, Germany
| | - Fedor Heidenreich
- Dept. of Neurology and Clinical Neurophysiology, Diakovere Krankenhaus Henriettenstift, Hannover, Germany
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
24
|
Chun MY, Jang H, Kim HJ, Kim JP, Gallacher J, Allué JA, Sarasa L, Castillo S, Pascual-Lucas M, Na DL, Seo SW. Contribution of clinical information to the predictive performance of plasma β-amyloid levels for amyloid positron emission tomography positivity. Front Aging Neurosci 2023; 15:1126799. [PMID: 36998318 PMCID: PMC10044013 DOI: 10.3389/fnagi.2023.1126799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/24/2023] [Indexed: 03/15/2023] Open
Abstract
BackgroundEarly detection of β-amyloid (Aβ) accumulation, a major biomarker for Alzheimer’s disease (AD), has become important. As fluid biomarkers, the accuracy of cerebrospinal fluid (CSF) Aβ for predicting Aβ deposition on positron emission tomography (PET) has been extensively studied, and the development of plasma Aβ is beginning to receive increased attention recently. In the present study, we aimed to determine whether APOE genotypes, age, and cognitive status increase the predictive performance of plasma Aβ and CSF Aβ levels for Aβ PET positivity.MethodsWe recruited 488 participants who underwent both plasma Aβ and Aβ PET studies (Cohort 1) and 217 participants who underwent both cerebrospinal fluid (CSF) Aβ and Aβ PET studies (Cohort 2). Plasma and CSF samples were analyzed using ABtest-MS, an antibody-free liquid chromatography-differential mobility spectrometry-triple quadrupole mass spectrometry method and INNOTEST enzyme-linked immunosorbent assay kits, respectively. To evaluate the predictive performance of plasma Aβ and CSF Aβ, respectively, logistic regression and receiver operating characteristic analyses were performed.ResultsWhen predicting Aβ PET status, both plasma Aβ42/40 ratio and CSF Aβ42 showed high accuracy (plasma Aβ area under the curve (AUC) 0.814; CSF Aβ AUC 0.848). In the plasma Aβ models, the AUC values were higher than plasma Aβ alone model, when the models were combined with either cognitive stage (p < 0.001) or APOE genotype (p = 0.011). On the other hand, there was no difference between the CSF Aβ models, when these variables were added.ConclusionPlasma Aβ might be a useful predictor of Aβ deposition on PET status as much as CSF Aβ, particularly when considered with clinical information such as APOE genotype and cognitive stage.
Collapse
Affiliation(s)
- Min Young Chun
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Republic of Korea
| | - Hyemin Jang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- *Correspondence: Hyemin Jang, ; Sang Won Seo,
| | - Hee Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jun Pyo Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Neuroimaging, Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John Gallacher
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Duk L. Na
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- *Correspondence: Hyemin Jang, ; Sang Won Seo,
| | | |
Collapse
|
25
|
Nirmalraj PN, Schneider T, Lüder L, Felbecker A. Protein fibril length in cerebrospinal fluid is increased in Alzheimer's disease. Commun Biol 2023; 6:251. [PMID: 36890343 PMCID: PMC9995532 DOI: 10.1038/s42003-023-04606-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/16/2023] [Indexed: 03/10/2023] Open
Abstract
Alzheimer's disease (AD) associated proteins exist in cerebrospinal fluid (CSF). This paper evidences that protein aggregate morphology distinctly differs in CSF of patients with AD dementia (ADD), mild cognitive impairment due to AD (MCI AD), with subjective cognitive decline without amyloid pathology (SCD) and with non-AD MCI using liquid-based atomic force microscopy (AFM). Spherical-shaped particles and nodular-shaped protofibrils were present in the CSF of SCD patients, whereas CSF of ADD patients abundantly contained elongated mature fibrils. Quantitative analysis of AFM topographs confirms fibril length is higher in CSF of ADD than in MCI AD and lowest in SCD and non-AD dementia patients. CSF fibril length is inversely correlated with CSF amyloid beta (Aβ) 42/40 ratio and CSF p-tau protein levels (obtained from biochemical assays) to predict amyloid and tau pathology with an accuracy of 94% and 82%, respectively, thus identifying ultralong protein fibrils in CSF as a possible signature of AD pathology.
Collapse
Affiliation(s)
- Peter Niraj Nirmalraj
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf, CH-8600, Switzerland.
| | - Thomas Schneider
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen, CH-9007, Switzerland
| | - Lars Lüder
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf, CH-8600, Switzerland
| | - Ansgar Felbecker
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen, CH-9007, Switzerland.
| |
Collapse
|
26
|
Cullen NC, Janelidze S, Mattsson-Carlgren N, Palmqvist S, Bittner T, Suridjan I, Jethwa A, Kollmorgen G, Brum WS, Zetterberg H, Blennow K, Stomrud E, Hansson O. Test-retest variability of plasma biomarkers in Alzheimer's disease and its effects on clinical prediction models. Alzheimers Dement 2023; 19:797-806. [PMID: 35699240 PMCID: PMC9747985 DOI: 10.1002/alz.12706] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/22/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The effect of random error on the performance of blood-based biomarkers for Alzheimer's disease (AD) must be determined before clinical implementation. METHODS We measured test-retest variability of plasma amyloid beta (Aβ)42/Aβ40, neurofilament light (NfL), glial fibrillary acidic protein (GFAP), and phosphorylated tau (p-tau)217 and simulated effects of this variability on biomarker performance when predicting either cerebrospinal fluid (CSF) Aβ status or conversion to AD dementia in 399 non-demented participants with cognitive symptoms. RESULTS Clinical performance was highest when combining all biomarkers. Among single-biomarkers, p-tau217 performed best. Test-retest variability ranged from 4.1% (Aβ42/Aβ40) to 25% (GFAP). This variability reduced the performance of the biomarkers (≈ΔAUC [area under the curve] -1% to -4%) with the least effects on models with p-tau217. The percent of individuals with unstable predicted outcomes was lowest for the multi-biomarker combination (14%). DISCUSSION Clinical prediction models combining plasma biomarkers-particularly p-tau217-exhibit high performance and are less effected by random error. Individuals with unstable predicted outcomes ("gray zone") should be recommended for further tests.
Collapse
Affiliation(s)
- Nicholas C. Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | | | | | | | - Wagner S. Brum
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, U.K
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for 27 Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
27
|
Constantinides VC, Paraskevas GP, Boufidou F, Bourbouli M, Pyrgelis ES, Stefanis L, Kapaki E. CSF Aβ42 and Aβ42/Aβ40 Ratio in Alzheimer's Disease and Frontotemporal Dementias. Diagnostics (Basel) 2023; 13:diagnostics13040783. [PMID: 36832271 PMCID: PMC9955886 DOI: 10.3390/diagnostics13040783] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Alzheimer's disease dementia (ADD) may manifest with atypical phenotypes, resembling behavioral variant frontotemporal dementia (bvFTD) and corticobasal syndrome (CBS), phenotypes which typically have an underlying frontotemporal lobar degeneration with tau proteinopathy (FTLD-tau), such as Pick's disease, corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), or FTLD with TDP-43 proteinopathy (FTLD-TDP). CSF biomarkers total and phosphorylated tau (τT and τP-181), and amyloid beta with 42 and 40 amino acids (Aβ42 and Aβ40) are biomarkers of AD pathology. The primary aim of this study was to compare the diagnostic accuracy of Aβ42 to Aβ42/Aβ40 ratio in: (a) differentiating ADD vs. frontotemporal dementias; (b) patients with AD pathology vs. non-AD pathologies; (c) compare biomarker ratios and composite markers to single CSF biomarkers in the differentiation of AD from FTD; Methods: In total, 263 subjects were included (ADD: n = 98; bvFTD: n = 49; PSP: n = 50; CBD: n = 45; controls: n = 21). CSF biomarkers were measured by commercially available ELISAs (EUROIMMUN). Multiple biomarker ratios (Aβ42/Aβ40; τT/τP-181; τT/Aβ42; τP-181/Aβ42) and composite markers (t-tau: τT/(Aβ42/Aβ40); p-tau: τP-181/(Aβ42/Aβ40) were calculated. ROC curve analysis was performed to compare AUCs of Aβ42 and Aβ42/Aβ40 ratio and relevant composite markers between ADD and FTD, as defined clinically. BIOMARKAPD/ABSI criteria (abnormal τT, τP-181 Aβ42, and Aβ42/Aβ40 ratio) were used to re-classify all patients into AD pathology vs. non-AD pathologies, and ROC curve analysis was repeated to compare Aβ42 and Aβ42/Aβ40; Results: Aβ42 did not differ from Aβ42/Aβ40 ratio in the differentiation of ADD from FTD (AUCs 0.752 and 0.788 respectively; p = 0.212). The τT/Aβ42 ratio provided maximal discrimination between ADD and FTD (AUC:0.893; sensitivity 88.8%, specificity 80%). BIOMARKAPD/ABSI criteria classified 60 patients as having AD pathology and 211 as non-AD. A total of 22 had discrepant results and were excluded. Aβ42/Aβ40 ratio was superior to Aβ42 in the differentiation of AD pathology from non-AD pathology (AUCs: 0.939 and 0.831, respectively; p < 0.001). In general, biomarker ratios and composite markers were superior to single CSF biomarkers in both analyses. CONCLUSIONS Aβ42/Aβ40 ratio is superior to Aβ42 in identifying AD pathology, irrespective of the clinical phenotype. CSF biomarker ratios and composite markers provide higher diagnostic accuracy compared to single CSF biomarkers.
Collapse
Affiliation(s)
- Vasilios C. Constantinides
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Correspondence: ; Tel.: +30-21-0728-9285
| | - George P. Paraskevas
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Second Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, Rimini 1, 12462 Athens, Greece
| | - Fotini Boufidou
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Mara Bourbouli
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Efstratios-Stylianos Pyrgelis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Leonidas Stefanis
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| | - Elisabeth Kapaki
- First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
- Neurochemistry and Biological Markers Unit, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, Vass. Sophias Ave. 74, 11528 Athens, Greece
| |
Collapse
|
28
|
Zou Y, Yu S, Ma X, Ma C, Mao C, Mu D, Li L, Gao J, Qiu L. How far is the goal of applying β-amyloid in cerebrospinal fluid for clinical diagnosis of Alzheimer's disease with standardization of measurements? Clin Biochem 2023; 112:33-42. [PMID: 36473516 DOI: 10.1016/j.clinbiochem.2022.11.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/02/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Cerebrospinal fluid (CSF) β-amyloid (Aβ) is important for early diagnosis of Alzheimer's disease (AD). However, the cohort distributions and cut-off values have large variation across different analytical assays, kits, and laboratories. In this review, we summarize the cut-off values and diagnostic performance for CSF Aβ1-42 and Aβ1-42/Aβ1-40, and explore the important effect factors. Based on the Alzheimer's Association external quality control program (AAQC program), the peer group coefficient of variation of manual ELISA assays for CSF Aβ1-42 was unsatisfied (>20%). Fully automated platforms with better performance have recently been developed, but still not widely applied. In 2020, the certified reference material (CRM) for CSF Aβ1-42 was launched; however, the AAQC 2021-round results did not show effective improvements. Thus, further development and popularization of CRM for CSF Aβ1-42 and Aβ1-40 are urgently required. Standardizing the diagnostic procedures of AD and related status and the pre-analytical protocols of CSF samples, improving detection performance of analytical assays, and popularizing the application of fully automated platforms are also important for the establishment of uniform cut-off values. Moreover, each laboratory should verify the applicability of uniform cut-off values, and evaluate whether it is necessary to establish its own population- and assay-specific cut-off values.
Collapse
Affiliation(s)
- Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Songlin Yu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Xiaoli Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China; Medical Science Research Center, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Chaochao Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Chenhui Mao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Danni Mu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Lei Li
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China
| | - Jing Gao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
29
|
Glans I, Sonestedt E, Nägga K, Gustavsson AM, González-Padilla E, Borne Y, Stomrud E, Melander O, Nilsson PM, Palmqvist S, Hansson O. Association Between Dietary Habits in Midlife With Dementia Incidence Over a 20-Year Period. Neurology 2023; 100:e28-e37. [PMID: 36224029 PMCID: PMC9827131 DOI: 10.1212/wnl.0000000000201336] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 08/16/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Dementia cases are expected to triple during the next 30 years, highlighting the importance of finding modifiable risk factors for dementia. The aim of this study was to investigate whether adherence to conventional dietary recommendations or to a modified Mediterranean diet are associated with a subsequent lower risk of developing all-cause dementia, Alzheimer disease (AD), vascular dementia (VaD), or with future accumulation of AD-related β-amyloid (Aβ) pathology. METHODS Baseline examination in the prospective Swedish population-based Malmö Diet and Cancer Study took place in 1991-1996 with a follow-up for incident dementia until 2014. Nondemented individuals born 1923-1950 and living in Malmö were invited to participate. Thirty thousand four hundred forty-six were recruited (41% of all eligible). Twenty-eight thousand twenty-five had dietary data and were included in this study. Dietary habits were assessed with a 7-day food diary, detailed food frequency questionnaire, and 1-hour interview. Main outcomes were incident all-cause dementia, AD, or VaD determined by memory clinic physicians. Secondary outcome was Aβ-accumulation measured using CSF Aβ42 (n = 738). Cox proportional hazard models were used to examine associations between diet and risk of developing dementia (adjusted for demographics, comorbidities, smoking, physical activity, and alcohol). RESULTS Sixty-one percent were women, and the mean (SD) age was 58.1 (7.6) years. One thousand nine hundred forty-three (6.9%) were diagnosed with dementia (median follow-up, 19.8 years). Individuals adhering to conventional dietary recommendations did not have lower risk of developing all-cause dementia (hazard ratio [HR] comparing worst with best adherence, 0.93, 95% CI 0.81-1.08), AD (HR 1.03, 0.85-1.23), or VaD (HR 0.93, 0.69-1.26). Neither did adherence to the modified Mediterranean diet lower the risk of developing all-cause dementia (HR 0.93 0.75-1.15), AD (HR 0.90, 0.68-1.19), or VaD (HR 1.00, 0.65-1.55). The results were similar when excluding participants developing dementia within 5 years or those with diabetes. No significant associations were found between diet and abnormal Aβ accumulation, conventional recommendations (OR 1.28, 0.74-2.24) or modified Mediterranean diet (OR 0.85, 0.39-1.84). DISCUSSION In this 20-year follow-up study, neither adherence to conventional dietary recommendations nor to modified Mediterranean diet were significantly associated with subsequent reduced risk for developing all-cause dementia, AD dementia, VaD, or AD pathology.
Collapse
Affiliation(s)
- Isabelle Glans
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden.
| | - Emily Sonestedt
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Katarina Nägga
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Anna-Märta Gustavsson
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Esther González-Padilla
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Yan Borne
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Olle Melander
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Peter M Nilsson
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit (I.G., K.N., A.-M.G., Erik Stomrud, S.P., O.H.), Department of Clinical Sciences Malmö, Lund University, Sweden; Memory Clinic (I.G., A.-M.G., Erik Stomrud, S.P., O.H.), Skåne University Hospital, Malmö, Sweden; Department of Clinical Sciences in Malmö(Emily Sonestedt, E.G.-P., Y.B.), Nutritional Epidemiology, Lund University, Skåne University Hospital, Malmö, Sweden; Department of Acute Internal Medicine and Geriatrics (K.N.), Linköping University, Sweden; and Department of Clinical Sciences Malmö (O.M., P.M.N.), Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
30
|
Liu L, Lauro BM, He A, Lee H, Bhattarai S, Wolfe MS, Bennett DA, Karch CM, Young-Pearse T, Selkoe DJ. Identification of the Aβ37/42 peptide ratio in CSF as an improved Aβ biomarker for Alzheimer's disease. Alzheimers Dement 2023; 19:79-96. [PMID: 35278341 PMCID: PMC9464800 DOI: 10.1002/alz.12646] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Identifying CSF-based biomarkers for the β-amyloidosis that initiates Alzheimer's disease (AD) could provide inexpensive and dynamic tests to distinguish AD from normal aging and predict future cognitive decline. METHODS We developed immunoassays specifically detecting all C-terminal variants of secreted amyloid β-protein and identified a novel biomarker, the Aβ 37/42 ratio, that outperforms the canonical Aβ42/40 ratio as a means to evaluate the γ-secretase activity and brain Aβ accumulation. RESULTS We show that Aβ 37/42 can distinguish physiological and pathological status in (1) presenilin-1 mutant vs wild-type cultured cells, (2) AD vs control brain tissue, and (3) AD versus cognitively normal (CN) subjects in CSF, where 37/42 (AUC 0.9622) outperformed 42/40 (AUC 0.8651) in distinguishing CN from AD. DISCUSSION We conclude that the Aβ 37/42 ratio sensitively detects presenilin/γ-secretase dysfunction and better distinguishes CN from AD than Aβ42/40 in CSF. Measuring this novel ratio alongside promising phospho-tau analytes may provide highly discriminatory fluid biomarkers for AD.
Collapse
Affiliation(s)
- Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Bianca M. Lauro
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Amy He
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Hyo Lee
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Sanjay Bhattarai
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS USA
| | - Michael S. Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center Rush University Medical Center, Chicago, IL USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St. Louis, MO USA
- Hope Center for Neurologic Disorders, St. Louis, MO USA
| | - Tracy Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | | | - Dennis J. Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
31
|
Yamashita K, Miura M, Watanabe S, Ishiki K, Arimatsu Y, Kawahira J, Kubo T, Sasaki K, Arai T, Hagino K, Irino Y, Nagai K, Verbel D, Koyama A, Dhadda S, Niiro H, Iwanaga S, Sato T, Yoshida T, Iwata A. Fully automated and highly specific plasma β-amyloid immunoassays predict β-amyloid status defined by amyloid positron emission tomography with high accuracy. Alzheimers Res Ther 2022; 14:86. [PMID: 35739591 PMCID: PMC9219197 DOI: 10.1186/s13195-022-01029-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/15/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Clinicians, researchers, and patients alike would greatly benefit from more accessible and inexpensive biomarkers for neural β-amyloid (Aβ). We aimed to assess the performance of fully automated plasma Aβ immunoassays, which correlate significantly with immunoprecipitation mass spectrometry assays, in predicting brain Aβ status as determined by visual read assessment of amyloid positron emission tomography (PET).
Methods
The plasma Aβ42/Aβ40 ratio was measured using a fully automated immunoassay platform (HISCL series) in two clinical studies (discovery and validation studies). The discovery and validation sample sets were retrospectively and randomly selected from participants with early Alzheimer’s disease (AD) identified during screening for the elenbecestat Phase 3 program.
Results
We included 197 participants in the discovery study (mean [SD] age 71.1 [8.5] years; 112 females) and 200 in the validation study (age 70.8 [7.9] years; 99 females). The plasma Aβ42/Aβ40 ratio predicted amyloid PET visual read status with areas under the receiver operating characteristic curves of 0.941 (95% confidence interval [CI] 0.910–0.973) and 0.868 (95% CI 0.816–0.920) in the discovery and validation studies, respectively. In the discovery study, a cutoff value of 0.102 was determined based on maximizing the Youden Index, and the sensitivity and specificity were calculated to be 96.0% (95% CI 90.1–98.9%) and 83.5% (95% CI 74.6–90.3%), respectively. Using the same cutoff value, the sensitivity and specificity in the validation study were calculated to be 88.0% (95% CI 80.0–93.6%) and 72.0% (95% CI 62.1–80.5%), respectively.
Conclusions
The plasma Aβ42/Aβ40 ratio measured using the HISCL series achieved high accuracy in predicting amyloid PET status. Since our blood-based immunoassay system is less invasive and more accessible than amyloid PET and cerebrospinal fluid testing, it may contribute to the diagnosis of AD in routine clinical practice.
Collapse
|
32
|
Luigi L, Silvia I, Viktor W, Wink AM, Mutsaerts HJMM, Sven H, Kaj B, O'Brien JT, Giovanni FB, Gael C, Pierre P, Pablo ML, Adam W, Joanna W, Craig R, Gispert JD, Visser PJ, Philip S, Frederik B, Tijms BM. Gray matter network properties show distinct associations with CSF p-tau 181 levels and amyloid status in individuals without dementia. AGING BRAIN 2022; 2:100054. [PMID: 36908898 PMCID: PMC9997148 DOI: 10.1016/j.nbas.2022.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/21/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Gray matter networks are altered with amyloid accumulation in the earliest stage of AD, and are associated with decline throughout the AD spectrum. It remains unclear to what extent gray matter network abnormalities are associated with hyperphosphorylated-tau (p-tau). We studied the relationship of cerebrospinal fluid (CSF) p-tau181 with gray matter networks in non-demented participants from the European Prevention of Alzheimer's Dementia (EPAD) cohort, and studied dependencies on amyloid and cognitive status. Gray matter networks were extracted from baseline structural 3D T1w MRI. P-tau181 and abeta were measured with the Roche cobas Elecsys System. We studied the associations of CSF biomarkers levels with several network's graph properties. We further studied whether the relationships of p-tau 181 and network measures were dependent on amyloid status and cognitive stage (CDR). We repeated these analyses for network properties at a regional level, where we averaged local network values across cubes within each of 116 areas as defined by the automated anatomical labeling (AAL) atlas. Amyloid positivity was associated with higher network size and betweenness centrality, and lower gamma, clustering and small-world coefficients. Higher CSF p-tau 181 levels were related to lower betweenness centrality, path length and lambda coefficients (all p < 0.01). Three-way interactions between p-tau181, amyloid status and CDR were found for path length, lambda and clustering (all p < 0.05): Cognitively unimpaired amyloid-negative participants showed lower path length and lambda values with higher CSF p-tau181 levels. Amyloid-positive participants with impaired cognition demonstrated lower clustering coefficients in association to higher CSF p-tau181 levels. Our results suggest that alterations in gray matter network clustering coefficient is an early and specific event in AD.
Collapse
Affiliation(s)
- Lorenzini Luigi
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Ingala Silvia
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Wottschel Viktor
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Alle Meije Wink
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Henk JMM Mutsaerts
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Ghent Institute for Functional and Metabolic Imaging (GIfMI), Ghent University, Ghent, Belgium
| | - Haller Sven
- CIMC – Centre d’Imagerie Médicale de Cornavin, Place de Cornavin 18, 1201 Genève, Switzerland
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, PR China
| | - Blennow Kaj
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - John T. O'Brien
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge School of Clinical Medicine, Box 189, Cambridge CB2 0QQ, UK
| | - Frisoni B. Giovanni
- Laboratory Alzheimer’s Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
- University Hospitals and University of Geneva, Geneva, Switzerland
| | - Chételat Gael
- Université de Normandie, Unicaen, Inserm, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders”, Institut Blood-and-Brain @ Caen-Normandie, Cyceron, 14000 Caen, France
| | - Payoux Pierre
- Department of Nuclear Medicine, Toulouse CHU, Purpan University Hospital, Toulouse, France
- Toulouse NeuroImaging Center, University of Toulouse, INSERM, UPS, Toulouse, France
| | - Martinez-Lage Pablo
- Centro de Investigación y Terapias Avanzadas, Neurología, CITA‐Alzheimer Foundation, San Sebastián, Spain
| | - Waldman Adam
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Wardlaw Joanna
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Centre at the University of Edinburgh, University of Edinburgh, UK
| | - Ritchie Craig
- Centre for Dementia Prevention, The University of Edinburgh, Scotland, UK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- CIBER Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Pieter Jelle Visser
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Alzheimer Center Limburg, Department of Psychiatry & Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Scheltens Philip
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Barkhof Frederik
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Institute of Neurology and Healthcare Engineering, University College London, London, UK
| | | |
Collapse
|
33
|
Xu C, Zhao L, Dong C. A Review of Application of Aβ42/40 Ratio in Diagnosis and Prognosis of Alzheimer’s Disease. J Alzheimers Dis 2022; 90:495-512. [DOI: 10.3233/jad-220673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The number of patients with Alzheimer’s disease (AD) and non-Alzheimer’s disease (non-AD) has drastically increased over recent decades. The amyloid cascade hypothesis attributes a vital role to amyloid-β protein (Aβ) in the pathogenesis of AD. As the main pathological hallmark of AD, amyloid plaques consist of merely the 42 and 40 amino acid variants of Aβ (Aβ 42 and Aβ 40). The cerebrospinal fluid (CSF) biomarker Aβ 42/40 has been extensively investigated and eventually integrated into important diagnostic tools to support the clinical diagnosis of AD. With the development of highly sensitive assays and technologies, blood-based Aβ 42/40, which was obtained using a minimally invasive and cost-effective method, has been proven to be abnormal in synchrony with CSF biomarker values. This paper presents the recent progress of the CSF Aβ 42/40 ratio and plasma Aβ 42/40 for AD as well as their potential clinical application as diagnostic markers or screening tools for dementia.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
34
|
Puig-Pijoan A, García-Escobar G, Fernández-Lebrero A, Manero Borràs R, Sánchez-Benavides G, Navalpotro-Gómez I, Cascales Lahoz D, Suárez-Calvet M, Grau-Rivera O, Boltes Alandí A, Pont-Sunyer M, Ortiz-Gil J, Carrillo-Molina S, López-Villegas D, Abellán-Vidal M, Martínez-Casamitjana M, Hernández-Sánchez J, Peña-Casanova J, Roquer J, Padrós Fluvià A, Puente-Périz V. Estudio CORCOBIA: determinación de puntos de corte de biomarcadores de enfermedad de Alzheimer en LCR en una cohorte clínica. Neurologia 2022. [DOI: 10.1016/j.nrl.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
35
|
Zetterberg H. Biofluid-based biomarkers for Alzheimer's disease-related pathologies: An update and synthesis of the literature. Alzheimers Dement 2022; 18:1687-1693. [PMID: 35213777 PMCID: PMC9514308 DOI: 10.1002/alz.12618] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/05/2021] [Accepted: 01/10/2022] [Indexed: 01/24/2023]
Abstract
The past few years have seen an explosion in sensitive and specific assays for cerebrospinal fluid (CSF) and blood biomarkers for Alzheimer's disease (AD) and related disorders, as well as some novel assays based on pathological seed-induced protein misfolding in patient samples. Here, I review this exciting field that promises to transform dementia diagnostics and disease monitoring. I discuss data on biomarkers for amyloid beta (Aβ) and tau pathology, neurodegeneration, and glial activation, mention the most promising biomarkers for α-synuclein and TDP-43 pathology, and highlight the need for further research into common co-pathologies. Finally, I consider practical aspects of blood-based biomarker-supported AD diagnostics and emphasize the importance of biomarker interpretation in a full clinical context.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| |
Collapse
|
36
|
Tippett LJ, Cawston EE, Morgan CA, Melzer TR, Brickell KL, Ilse C, Cheung G, Kirk IJ, Roberts RP, Govender J, Griner L, Le Heron C, Buchanan S, Port W, Dudley M, Anderson TJ, Williams JM, Cutfield NJ, Dalrymple-Alford JC, Wood P. Dementia Prevention Research Clinic: a longitudinal study investigating factors influencing the development of Alzheimer's disease in Aotearoa, New Zealand. J R Soc N Z 2022; 53:489-510. [PMID: 39439970 PMCID: PMC11459802 DOI: 10.1080/03036758.2022.2098780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/04/2022] [Indexed: 10/15/2022]
Abstract
Aotearoa New Zealand's population is ageing. Increasing life expectancy is accompanied by increases in prevalence of Alzheimer's Disease (AD) and ageing-related disorders. The multicentre Dementia Prevention Research Clinic longitudinal study aims to improve understanding of AD and dementia in Aotearoa, in order to develop interventions that delay or prevent progression to dementia. Comprising research clinics in Auckland, Christchurch and Dunedin, this multi-disciplinary study involves community participants who undergo biennial investigations informed by international protocols and best practice: clinical, neuropsychological, neuroimaging, lifestyle evaluations, APOE genotyping, blood collection and processing. A key research objective is to identify a 'biomarker signature' that predicts progression from mild cognitive impairment to AD. Candidate biomarkers include: blood proteins and microRNAs, genetic, neuroimaging and neuropsychological markers, health, cultural, lifestyle, sensory and psychosocial factors. We are examining a range of mechanisms underlying the progression of AD pathology (e.g. faulty blood-brain barrier, excess parenchymal iron, vascular dysregulation). This paper will outline key aspects of the Dementia Prevention Research Clinic's research, provide an overview of data collection, and a summary of 266 participants recruited to date. The national outreach of the clinics is a strength; the heart of the Dementia Prevention Research Clinics are its people.
Collapse
Affiliation(s)
- Lynette J. Tippett
- NZ-Dementia Prevention Research Clinic, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Erin E. Cawston
- NZ-Dementia Prevention Research Clinic, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Catherine A. Morgan
- NZ-Dementia Prevention Research Clinic, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Tracy R. Melzer
- NZ-Dementia Prevention Research Clinic, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Kiri L. Brickell
- NZ-Dementia Prevention Research Clinic, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- School of Medicine, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Christina Ilse
- NZ-Dementia Prevention Research Clinic, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Gary Cheung
- NZ-Dementia Prevention Research Clinic, New Zealand
- Department of Psychological Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Ian J. Kirk
- NZ-Dementia Prevention Research Clinic, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Reece P. Roberts
- NZ-Dementia Prevention Research Clinic, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Jane Govender
- NZ-Dementia Prevention Research Clinic, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Leon Griner
- NZ-Dementia Prevention Research Clinic, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Campbell Le Heron
- NZ-Dementia Prevention Research Clinic, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
- Dept of Neurology, Canterbury District Health Board, Christchurch, New Zealand
| | - Sarah Buchanan
- NZ-Dementia Prevention Research Clinic, New Zealand
- Department of Neurology, Southern District Health Board, Dunedin, New Zealand
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Waiora Port
- NZ-Dementia Prevention Research Clinic, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Makarena Dudley
- NZ-Dementia Prevention Research Clinic, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Tim J. Anderson
- NZ-Dementia Prevention Research Clinic, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
- Dept of Neurology, Canterbury District Health Board, Christchurch, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Joanna M. Williams
- NZ-Dementia Prevention Research Clinic, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Nicholas J. Cutfield
- NZ-Dementia Prevention Research Clinic, New Zealand
- Department of Neurology, Southern District Health Board, Dunedin, New Zealand
- Department of Medicine, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - John C. Dalrymple-Alford
- NZ-Dementia Prevention Research Clinic, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - Phil Wood
- NZ-Dementia Prevention Research Clinic, New Zealand
- School of Medicine, University of Auckland, Auckland, New Zealand
- Ministry of Health, Wellington, New Zealand
- Department of Older Adults and Home Health, Waitemata District Health Board, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, Dunedin, New Zealand
| | - the NZ-DPRC
- NZ-Dementia Prevention Research Clinic, New Zealand
| |
Collapse
|
37
|
Han J, Feng GH, Liu HW, Yi JP, Wu JB, Yao XX. Classifying mild cognitive impairment and Alzheimer's disease by constructing a 14-gene diagnostic model. Am J Transl Res 2022; 14:4477-4492. [PMID: 35958496 PMCID: PMC9360837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and mild cognitive impairment (MCI) are two neurodegenerative diseases. Most patients with MCI will develop AD. Early detection of AD and MCI is a crucial issue in terms of secondary prevention. Therefore, more diagnostic models need to be developed to distinguish AD patients from MCI patients. METHODS In our research, the expression matrix and were screened from Gene Expression Omnibus (GEO) databases. A 14-gene diagnostic model was constructed with lasso logistic analysis. The efficiency and accuracy of diagnostic model have also been validated. In order to clarify the expression differences of 14 genes in health donor, AD and MCI, the blood samples of patients and healthy individuals were collected. The mRNA expression of the 14 genes in blood sample were detected. The SH-SY5Y cell injury model was constructed and biological function of POU2AF1 and ANKRD22 in SH-SY5Y have been proved. RESULTS We obtained 16 genes which have an area under curve (AUC) ≥0.6. After that, a diagnostic model based on 14 genes was constructed. Validation in independent cohort showed that the diagnostic model has a good diagnostic efficiency. The expressions of 6 genes in AD patients were significantly lower than those in healthy individuals and MCI patients, while the expressions of 8 genes in AD patients were significantly higher than those in healthy individuals and MCI patients. In in vitro experiments, we found that two key genes POU2AF1 and ANKRD22 could regulate neuronal development by regulating cell viability and IL-6 expression. CONCLUSION The diagnostic model established in this study has a good diagnose efficiency. Most of these genes in diagnostic model also showed diagnostic value in AD patients. This research also can help doctors make better diagnosis for the treatment and prevention of AD.
Collapse
Affiliation(s)
- Jing Han
- School of Basic Medical Sciences, Xiangnan UniversityChenzhou 423000, Hunan, China
| | - Gang-Hua Feng
- Department of Neurology, Chenzhou First People’s HospitalChenzhou 423000, Hunan, China
| | - Hua-Wu Liu
- School of Basic Medical Sciences, Xiangnan UniversityChenzhou 423000, Hunan, China
| | - Ji-Ping Yi
- Department of Neurology, Chenzhou First People’s HospitalChenzhou 423000, Hunan, China
| | - Ji-Bao Wu
- Department of Neurology, Chenzhou First People’s HospitalChenzhou 423000, Hunan, China
| | - Xiao-Xi Yao
- Department of Neurology, Chenzhou First People’s HospitalChenzhou 423000, Hunan, China
| |
Collapse
|
38
|
Chen YH, Lin RR, Huang HF, Xue YY, Tao QQ. Microglial Activation, Tau Pathology, and Neurodegeneration Biomarkers Predict Longitudinal Cognitive Decline in Alzheimer's Disease Continuum. Front Aging Neurosci 2022; 14:848180. [PMID: 35847667 PMCID: PMC9280990 DOI: 10.3389/fnagi.2022.848180] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/12/2022] [Indexed: 01/02/2023] Open
Abstract
Purpose Biomarkers used for predicting longitudinal cognitive change in Alzheimer's disease (AD) continuum are still elusive. Tau pathology, neuroinflammation, and neurodegeneration are the leading candidate predictors. We aimed to determine these three aspects of biomarkers in cerebrospinal fluid (CSF) and plasma to predict longitudinal cognition status using Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Patients and Methods A total of 430 subjects including, 96 cognitive normal (CN) with amyloid β (Aβ)-negative, 54 CN with Aβ-positive, 195 mild cognitive impairment (MCI) with Aβ-positive, and 85 AD with amyloid-positive (Aβ-positive are identified by CSF Aβ42/Aβ40 < 0.138). Aβ burden was evaluated by CSF and plasma Aβ42/Aβ40 ratio; tau pathology was evaluated by CSF and plasma phosphorylated-tau (p-tau181); microglial activation was measured by CSF soluble TREM2 (sTREM2) and progranulin (PGRN); neurodegeneration was measured by CSF and plasma t-tau and structural magnetic resonance imaging (MRI); cognition was examined annually over the subsequent 8 years using the Alzheimer's Disease Assessment Scale Cognition 13-item scale (ADAS13) and Mini-Mental State Exam (MMSE). Linear mixed-effects models (LME) were applied to assess the correlation between biomarkers and longitudinal cognition decline, as well as their effect size on the prediction of longitudinal cognitive decline. Results Baseline CSF Aβ42/Aβ40 ratio was decreased in MCI and AD compared to CN, while CSF p-tau181 and t-tau increased. Baseline CSF sTREM2 and PGRN did not show any differences in MCI and AD compared to CN. Baseline brain volumes (including the hippocampal, entorhinal, middle temporal lobe, and whole-brain) decreased in MCI and AD groups. For the longitudinal study, there were significant interaction effects of CSF p-tau181 × time, plasma p-tau181 × time, CSF sTREM2 × time, and brain volumes × time, indicating CSF, and plasma p-tau181, CSF sTREM2, and brain volumes could predict longitudinal cognition deterioration rate. CSF sTREM2, CSF, and plasma p-tau181 had similar medium prediction effects, while brain volumes showed stronger effects in predicting cognition decline. Conclusion Our study reported that baseline CSF sTREM2, CSF, and plasma p-tau181, as well as structural MRI, could predict longitudinal cognitive decline in subjects with positive AD pathology. Plasma p-tau181 can be used as a relatively noninvasive reliable biomarker for AD longitudinal cognition decline prediction.
Collapse
Affiliation(s)
- Yi-He Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Rong-Rong Lin
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui-Feng Huang
- Department of Neurology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Yan-Yan Xue
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing-Qing Tao
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
39
|
Orellana A, García-González P, Valero S, Montrreal L, de Rojas I, Hernández I, Rosende-Roca M, Vargas L, Tartari JP, Esteban-De Antonio E, Bojaryn U, Narvaiza L, Alarcón-Martín E, Alegret M, Alcolea D, Lleó A, Tárraga L, Pytel V, Cano A, Marquié M, Boada M, Ruiz A. Establishing In-House Cutoffs of CSF Alzheimer’s Disease Biomarkers for the AT(N) Stratification of the Alzheimer Center Barcelona Cohort. Int J Mol Sci 2022; 23:ijms23136891. [PMID: 35805894 PMCID: PMC9266894 DOI: 10.3390/ijms23136891] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Clinical diagnosis of Alzheimer’s disease (AD) increasingly incorporates CSF biomarkers. However, due to the intrinsic variability of the immunodetection techniques used to measure these biomarkers, establishing in-house cutoffs defining the positivity/negativity of CSF biomarkers is recommended. However, the cutoffs currently published are usually reported by using cross-sectional datasets, not providing evidence about its intrinsic prognostic value when applied to real-world memory clinic cases. Methods: We quantified CSF Aβ1-42, Aβ1-40, t-Tau, and p181Tau with standard INNOTEST® ELISA and Lumipulse G® chemiluminescence enzyme immunoassay (CLEIA) performed on the automated Lumipulse G600II. Determination of cutoffs included patients clinically diagnosed with probable Alzheimer’s disease (AD, n = 37) and subjective cognitive decline subjects (SCD, n = 45), cognitively stable for 3 years and with no evidence of brain amyloidosis in 18F-Florbetaben-labeled positron emission tomography (FBB-PET). To compare both methods, a subset of samples for Aβ1-42 (n = 519), t-Tau (n = 399), p181Tau (n = 77), and Aβ1-40 (n = 44) was analyzed. Kappa agreement of single biomarkers and Aβ1-42/Aβ1-40 was evaluated in an independent group of mild cognitive impairment (MCI) and dementia patients (n = 68). Next, established cutoffs were applied to a large real-world cohort of MCI subjects with follow-up data available (n = 647). Results: Cutoff values of Aβ1-42 and t-Tau were higher for CLEIA than for ELISA and similar for p181Tau. Spearman coefficients ranged between 0.81 for Aβ1-40 and 0.96 for p181TAU. Passing–Bablok analysis showed a systematic and proportional difference for all biomarkers but only systematic for Aβ1-40. Bland–Altman analysis showed an average difference between methods in favor of CLEIA. Kappa agreement for single biomarkers was good but lower for the Aβ1-42/Aβ1-40 ratio. Using the calculated cutoffs, we were able to stratify MCI subjects into four AT(N) categories. Kaplan–Meier analyses of AT(N) categories demonstrated gradual and differential dementia conversion rates (p = 9.815−27). Multivariate Cox proportional hazard models corroborated these findings, demonstrating that the proposed AT(N) classifier has prognostic value. AT(N) categories are only modestly influenced by other known factors associated with disease progression. Conclusions: We established CLEIA and ELISA internal cutoffs to discriminate AD patients from amyloid-negative SCD individuals. The results obtained by both methods are not interchangeable but show good agreement. CLEIA is a good and faster alternative to manual ELISA for providing AT(N) classification of our patients. AT(N) categories have an impact on disease progression. AT(N) classifiers increase the certainty of the MCI prognosis, which can be instrumental in managing real-world MCI subjects.
Collapse
Affiliation(s)
- Adelina Orellana
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Pablo García-González
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Sergi Valero
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Isabel Hernández
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Maitee Rosende-Roca
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Liliana Vargas
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Ester Esteban-De Antonio
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Urszula Bojaryn
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Leire Narvaiza
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Emilio Alarcón-Martín
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Montserrat Alegret
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Daniel Alcolea
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, 08029 Barcelona, Spain
| | - Alberto Lleó
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, 08029 Barcelona, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
| | - Amanda Cano
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Marta Marquié
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08029 Barcelona, Spain; (A.O.); (P.G.-G.); (S.V.); (L.M.); (I.d.R.); (I.H.); (M.R.-R.); (L.V.); (J.P.T.); (E.E.-D.A.); (U.B.); (L.N.); (E.A.-M.); (M.A.); (L.T.); (V.P.); (A.C.); (M.M.); (M.B.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; (D.A.); (A.L.)
- Correspondence:
| |
Collapse
|
40
|
Treatment effects on event-related EEG potentials and oscillations in Alzheimer's disease. Int J Psychophysiol 2022; 177:179-201. [PMID: 35588964 DOI: 10.1016/j.ijpsycho.2022.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease dementia (ADD) is the most diffuse neurodegenerative disorder belonging to mild cognitive impairment (MCI) and dementia in old persons. This disease is provoked by an abnormal accumulation of amyloid-beta and tauopathy proteins in the brain. Very recently, the first disease-modifying drug has been licensed with reserve (i.e., Aducanumab). Therefore, there is a need to identify and use biomarkers probing the neurophysiological underpinnings of human cognitive functions to test the clinical efficacy of that drug. In this regard, event-related electroencephalographic potentials (ERPs) and oscillations (EROs) are promising candidates. Here, an Expert Panel from the Electrophysiology Professional Interest Area of the Alzheimer's Association and Global Brain Consortium reviewed the field literature on the effects of the most used symptomatic drug against ADD (i.e., Acetylcholinesterase inhibitors) on ERPs and EROs in ADD patients with MCI and dementia at the group level. The most convincing results were found in ADD patients. In those patients, Acetylcholinesterase inhibitors partially normalized ERP P300 peak latency and amplitude in oddball paradigms using visual stimuli. In these same paradigms, those drugs partially normalize ERO phase-locking at the theta band (4-7 Hz) and spectral coherence between electrode pairs at the gamma (around 40 Hz) band. These results are of great interest and may motivate multicentric, double-blind, randomized, and placebo-controlled clinical trials in MCI and ADD patients for final cross-validation.
Collapse
|
41
|
Janelidze S, Palmqvist S, Leuzy A, Stomrud E, Verberk IMW, Zetterberg H, Ashton NJ, Pesini P, Sarasa L, Allué JA, Teunissen CE, Dage JL, Blennow K, Mattsson-Carlgren N, Hansson O. Detecting amyloid positivity in early Alzheimer's disease using combinations of plasma Aβ42/Aβ40 and p-tau. Alzheimers Dement 2022; 18:283-293. [PMID: 34151519 DOI: 10.1002/alz.12395] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 01/20/2023]
Abstract
INTRODUCTION We studied usefulness of combining blood amyloid beta (Aβ)42/Aβ40, phosphorylated tau (p-tau)217, and neurofilament light (NfL) to detect abnormal brain Aβ deposition in different stages of early Alzheimer's disease (AD). METHODS Plasma biomarkers were measured using mass spectrometry (Aβ42/Aβ40) and immunoassays (p-tau217 and NfL) in cognitively unimpaired individuals (CU, N = 591) and patients with mild cognitive impairment (MCI, N = 304) from two independent cohorts (BioFINDER-1, BioFINDER-2). RESULTS In CU, a combination of plasma Aβ42/Aβ40 and p-tau217 detected abnormal brain Aβ status with area under the curve (AUC) of 0.83 to 0.86. In MCI, the models including p-tau217 alone or Aβ42/Aβ40 and p-tau217 had similar AUCs (0.86-0.88); however, the latter showed improved model fit. The models were implemented in an online application providing individualized risk assessments (https://brainapps.shinyapps.io/PredictABplasma/). DISCUSSION A combination of plasma Aβ42/Aβ40 and p-tau217 discriminated Aβ status with relatively high accuracy, whereas p-tau217 showed strongest associations with Aβ pathology in MCI but not in CU.
Collapse
Affiliation(s)
- Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Skåne University Hospital, Malmö, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Skåne University Hospital, Malmö, Sweden
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | | | | | | | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
42
|
Bogolepova A, Makhnovich E, Kovalenko E, Osinovskaya N. Potential biomarkers of early diagnosis of Alzheimer’s disease. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:7-14. [DOI: 10.17116/jnevro20221220917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
43
|
Pavlik VN, Burnham SC, Kass JS, Helmer C, Palmqvist S, Vassilaki M, Dartigues JF, Hansson O, Masters CL, Pérès K, Petersen RC, Stomrud E, Butler L, Coloma PM, Teitsma XM, Doody R, Sano M. Connecting Cohorts to Diminish Alzheimer's Disease (CONCORD-AD): A Report of an International Research Collaboration Network. J Alzheimers Dis 2022; 85:31-45. [PMID: 34776434 PMCID: PMC8842789 DOI: 10.3233/jad-210525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 11/15/2022]
Abstract
Longitudinal observational cohort studies are being conducted worldwide to understand cognition, biomarkers, and the health of the aging population better. Cross-cohort comparisons and networks of registries in Alzheimer's disease (AD) foster scientific exchange, generate insights, and contribute to the evolving clinical science in AD. A scientific working group was convened with invited investigators from established cohort studies in AD, in order to form a research collaboration network as a resource to address important research questions. The Connecting Cohorts to Diminish Alzheimer's Disease (CONCORD-AD) collaboration network was created to bring together global resources and expertise, to generate insights and improve understanding of the natural history of AD, to inform design of clinical trials in all disease stages, and to plan for optimal patient access to disease-modifying therapies once they become available. The network brings together expertise and data insights from 7 cohorts across Australia, Europe, and North America. Notably, the network includes populations recruited through memory clinics as well as population-based cohorts, representing observations from individuals across the AD spectrum. This report aims to introduce the CONCORD-AD network, providing an overview of the cohorts involved, reporting the common assessments used, and describing the key characteristics of the cohort populations. Cohort study designs and baseline population characteristics are compared, and available cognitive, functional, and neuropsychiatric symptom data, as well as the frequency of biomarker assessments, are summarized. Finally, the challenges and opportunities of cross-cohort studies in AD are discussed.
Collapse
Affiliation(s)
- Valory N. Pavlik
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Samantha C. Burnham
- The Australian eHealth Research Centre, CSIRO Health and Biosecurity, Melbourne, VIC, Australia
| | - Joseph S. Kass
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Catherine Helmer
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Jean-François Dartigues
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
- Department of Neurology, Memory Consultation, Bordeaux University Hospital, Bordeaux, France
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Colin L. Masters
- The Florey Institute and The University of Melbourne, Parkville, VIC, Australia
| | - Karine Pérès
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
| | - Ronald C. Petersen
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Lesley Butler
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Preciosa M. Coloma
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Xavier M. Teitsma
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Rachelle Doody
- Product Development Neuroscience, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Product Development Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | - Mary Sano
- Department of Psychiatry, Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - for the CONCORD-AD investigators
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
- The Australian eHealth Research Centre, CSIRO Health and Biosecurity, Melbourne, VIC, Australia
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR, Bordeaux, France
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Memory Consultation, Bordeaux University Hospital, Bordeaux, France
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- The Florey Institute and The University of Melbourne, Parkville, VIC, Australia
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Product Development Personalised Health Care – Data Science, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Product Development Neuroscience, F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Product Development Neuroscience, Genentech, Inc., South San Francisco, CA, USA
- Department of Psychiatry, Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
44
|
Jiang C, Wang Q, Xie S, Chen Z, Fu L, Peng Q, Liang Y, Guo H, Guo T. OUP accepted manuscript. Brain Commun 2022; 4:fcac084. [PMID: 35441134 PMCID: PMC9014538 DOI: 10.1093/braincomms/fcac084] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/21/2021] [Accepted: 03/29/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Chenyang Jiang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Qingyong Wang
- Department of Neurology, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen 518107, China
| | - Siwei Xie
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Zhicheng Chen
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Liping Fu
- Department of Nuclear Medicine, China-Japan Friendship Hospital, 2 Yinghuayuan Dongjie, Beijing 100029, China
| | - Qiyu Peng
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Hongbo Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Correspondence to: Tengfei Guo, PhD Institute of Biomedical Engineering Shenzhen Bay Laboratory, No.5 Kelian Road Shenzhen 518132, China E-mail:
| | | |
Collapse
|
45
|
Ingala S, Tomassen J, Collij LE, Prent N, van 't Ent D, Ten Kate M, Konijnenberg E, Yaqub M, Scheltens P, de Geus EJC, Teunissen CE, Tijms B, Wink AM, Barkhof F, van Berckel BNM, Visser PJ, den Braber A. Amyloid-driven disruption of default mode network connectivity in cognitively healthy individuals. Brain Commun 2021; 3:fcab201. [PMID: 34617016 PMCID: PMC8490784 DOI: 10.1093/braincomms/fcab201] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/06/2021] [Accepted: 05/03/2021] [Indexed: 12/03/2022] Open
Abstract
Cortical accumulation of amyloid beta is one of the first events of Alzheimer’s disease pathophysiology, and has been suggested to follow a consistent spatiotemporal ordering, starting in the posterior cingulate cortex, precuneus and medio-orbitofrontal cortex. These regions overlap with those of the default mode network, a brain network also involved in memory functions. Aberrant default mode network functional connectivity and higher network sparsity have been reported in prodromal and clinical Alzheimer’s disease. We investigated the association between amyloid burden and default mode network connectivity in the preclinical stage of Alzheimer’s disease and its association with longitudinal memory decline. We included 173 participants, in which amyloid burden was assessed both in CSF by the amyloid beta 42/40 ratio, capturing the soluble part of amyloid pathology, and in dynamic PET scans calculating the non-displaceable binding potential in early-stage regions. The default mode network was identified with resting-state functional MRI. Then, we calculated functional connectivity in the default mode network, derived from independent component analysis, and eigenvector centrality, a graph measure recursively defining important nodes on the base of their connection with other important nodes. Memory was tested at baseline, 2- and 4-year follow-up. We demonstrated that higher amyloid burden as measured by both CSF amyloid beta 42/40 ratio and non-displaceable binding potential in the posterior cingulate cortex was associated with lower functional connectivity in the default mode network. The association between amyloid burden (CSF and non-displaceable binding potential in the posterior cingulate cortex) and aberrant default mode network connectivity was confirmed at the voxel level with both functional connectivity and eigenvector centrality measures, and it was driven by voxel clusters localized in the precuneus, cingulate, angular and left middle temporal gyri. Moreover, we demonstrated that functional connectivity in the default mode network predicts longitudinal memory decline synergistically with regional amyloid burden, as measured by non-displaceable binding potential in the posterior cingulate cortex. Taken together, these results suggest that early amyloid beta deposition is associated with aberrant default mode network connectivity in cognitively healthy individuals and that default mode network connectivity markers can be used to identify subjects at risk of memory decline.
Collapse
Affiliation(s)
- Silvia Ingala
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Naomi Prent
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands.,Faculty of Behavioral and Movement Sciences, Section Clinical Neuropsychology, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.,Vesalius, Centre for Neuropsychiatry, GGZ Altrecht, 3447 GM Woerden, The Netherlands
| | - Dennis van 't Ent
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Neuroscience Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Mara Ten Kate
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands.,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Elles Konijnenberg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Neuroscience Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - Betty Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Alle Meije Wink
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands.,Institute of Neurology and Healthcare Engineering, University College London, WC1E 6BT London, UK
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Location VUmc, 1081 HZ Amsterdam, The Netherlands.,Department of Biological Psychology, Vrije Universiteit Amsterdam, Neuroscience Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
46
|
Korecka M, Shaw LM. Mass spectrometry-based methods for robust measurement of Alzheimer's disease biomarkers in biological fluids. J Neurochem 2021; 159:211-233. [PMID: 34244999 PMCID: PMC9057379 DOI: 10.1111/jnc.15465] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/11/2021] [Accepted: 07/06/2021] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting 60%-70% of people afflicted with this disease. Accurate antemortem diagnosis is urgently needed for early detection of AD to enable reliable estimation of prognosis, intervention, and monitoring of the disease. The National Institute on Aging/Alzheimer's Association sponsored the 'Research Framework: towards a biological definition of AD', which recommends using different biomarkers in living persons for a biomarker-based definition of AD regardless of clinical status. Fluid biomarkers represent one of key groups of them. Since cerebrospinal fluid (CSF) is in direct contact with brain and many proteins present in the brain can be detected in CSF, this fluid has been regarded as the best biofluid in which to measure AD biomarkers. Recently, technological advancements in protein detection made possible the effective study of plasma AD biomarkers despite their significantly lower concentrations versus to that in CSF. This and other challenges that face plasma-based biomarker measurements can be overcome by using mass spectrometry. In this review, we discuss AD biomarkers which can be reliably measured in CSF and plasma using targeted mass spectrometry coupled to liquid chromatography (LC/MS/MS). We describe progress in LC/MS/MS methods' development, emphasize the challenges, and summarize major findings. We also highlight the role of mass spectrometry and progress made in the process of global standardization of the measurement of Aβ42/Aβ40. Finally, we briefly describe exploratory proteomics which seek to identify new biomarkers that can contribute to detection of co-pathological processes that are common in sporadic AD.
Collapse
Affiliation(s)
- Magdalena Korecka
- Department of Pathology and Laboratory Medicine Perlman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine Perlman School of Medicine University of Pennsylvania Philadelphia PA USA
| |
Collapse
|
47
|
Hampel H, Hardy J, Blennow K, Chen C, Perry G, Kim SH, Villemagne VL, Aisen P, Vendruscolo M, Iwatsubo T, Masters CL, Cho M, Lannfelt L, Cummings JL, Vergallo A. The Amyloid-β Pathway in Alzheimer's Disease. Mol Psychiatry 2021; 26:5481-5503. [PMID: 34456336 PMCID: PMC8758495 DOI: 10.1038/s41380-021-01249-0] [Citation(s) in RCA: 843] [Impact Index Per Article: 210.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Breakthroughs in molecular medicine have positioned the amyloid-β (Aβ) pathway at the center of Alzheimer's disease (AD) pathophysiology. While the detailed molecular mechanisms of the pathway and the spatial-temporal dynamics leading to synaptic failure, neurodegeneration, and clinical onset are still under intense investigation, the established biochemical alterations of the Aβ cycle remain the core biological hallmark of AD and are promising targets for the development of disease-modifying therapies. Here, we systematically review and update the vast state-of-the-art literature of Aβ science with evidence from basic research studies to human genetic and multi-modal biomarker investigations, which supports a crucial role of Aβ pathway dyshomeostasis in AD pathophysiological dynamics. We discuss the evidence highlighting a differentiated interaction of distinct Aβ species with other AD-related biological mechanisms, such as tau-mediated, neuroimmune and inflammatory changes, as well as a neurochemical imbalance. Through the lens of the latest development of multimodal in vivo biomarkers of AD, this cross-disciplinary review examines the compelling hypothesis- and data-driven rationale for Aβ-targeting therapeutic strategies in development for the early treatment of AD.
Collapse
Affiliation(s)
- Harald Hampel
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| | - John Hardy
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Christopher Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea; Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Aisen
- USC Alzheimer's Therapeutic Research Institute, San Diego, CA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Colin L Masters
- Laureate Professor of Dementia Research, Florey Institute and The University of Melbourne, Parkville, VIC, Australia
| | - Min Cho
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA
| | - Lars Lannfelt
- Uppsala University, Department of of Public Health/Geriatrics, Uppsala, Sweden
- BioArctic AB, Stockholm, Sweden
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Andrea Vergallo
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| |
Collapse
|
48
|
Grothe MJ, Moscoso A, Ashton NJ, Karikari TK, Lantero-Rodriguez J, Snellman A, Zetterberg H, Blennow K, Schöll M. Associations of Fully Automated CSF and Novel Plasma Biomarkers With Alzheimer Disease Neuropathology at Autopsy. Neurology 2021; 97:e1229-e1242. [PMID: 34266917 PMCID: PMC8480485 DOI: 10.1212/wnl.0000000000012513] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/24/2021] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE To study CSF biomarkers of Alzheimer disease (AD) analyzed by fully automated Elecsys immunoassays compared to neuropathologic gold standards and to compare their accuracy to plasma phosphorylated tau (p-tau181) measured with a novel single molecule array method. METHODS We studied antemortem Elecsys-derived CSF biomarkers in 45 individuals who underwent standardized postmortem assessments of AD and non-AD neuropathologic changes at autopsy. In a subset of 26 participants, we also analyzed antemortem levels of plasma p-tau181 and neurofilament light (NfL). Reference biomarker values were obtained from 146 amyloid-PET-negative healthy controls (HC). RESULTS All CSF biomarkers clearly distinguished pathology-confirmed AD dementia (n = 27) from HC (area under the curve [AUC] 0.86-1.00). CSF total tau (t-tau), p-tau181, and their ratios with β-amyloid1-42 (Aβ1-42) also accurately distinguished pathology-confirmed AD from non-AD dementia (n = 8; AUC 0.94-0.97). In pathology-specific analyses, intermediate to high Thal amyloid phases were best detected by CSF Aβ1-42 (AUC [95% confidence interval] 0.91 [0.81-1]), while intermediate to high scores for Consortium to Establish a Registry for Alzheimer's Disease neuritic plaques and Braak tau stages were best detected by CSF p-tau181 (AUC 0.89 [0.79-0.99] and 0.88 [0.77-0.99], respectively). Optimal Elecsys biomarker cutoffs were derived at 1,097, 229, and 19 pg/mL for Aβ1-42, t-tau, and p-tau181. In the plasma subsample, both plasma p-tau181 (AUC 0.91 [0.86-0.96]) and NfL (AUC 0.93 [0.87-0.99]) accurately distinguished those with pathology-confirmed AD (n = 14) from HC. However, only p-tau181 distinguished AD from non-AD dementia cases (n = 4; AUC 0.96 [0.88-1.00]) and showed a similar, although weaker, pathologic specificity for neuritic plaques (AUC 0.75 [0.52-0.98]) and Braak stage (AUC 0.71 [0.44-0.98]) as CSF p-tau181. CONCLUSION Elecsys-derived CSF biomarkers detect AD neuropathologic changes with very high discriminative accuracy in vivo. Preliminary findings support the use of plasma p-tau181 as an easily accessible and scalable biomarker of AD pathology. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that fully automated CSF t-tau and p-tau181 measurements discriminate between autopsy-confirmed AD and other dementias.
Collapse
Affiliation(s)
- Michel J Grothe
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK.
| | - Alexis Moscoso
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK
| | - Nicholas J Ashton
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK
| | - Thomas K Karikari
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK
| | - Juan Lantero-Rodriguez
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK
| | - Anniina Snellman
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK
| | - Henrik Zetterberg
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK
| | - Kaj Blennow
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK
| | - Michael Schöll
- From the Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Department of Psychiatry and Neurochemistry (M.J.G., A.M., N.J.A., T.K.K., J.L.-R., A.S., H.Z., K.B., M.S.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy, and Wallenberg Centre for Molecular and Translational Medicine (M.J.G., A.M., N.J.A., M.S.), University of Gothenburg, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), London, UK; Turku PET Centre (A.S.), University of Turku, Finland; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z., M.S.), UCL Institute of Neurology; and UK Dementia Research Institute at UCL (H.Z.), London, UK.
| |
Collapse
|
49
|
Lombardi G, Pupi A, Bessi V, Polito C, Padiglioni S, Ferrari C, Lucidi G, Berti V, De Cristofaro MT, Piaceri I, Bagnoli S, Nacmias B, Sorbi S. Challenges in Alzheimer's Disease Diagnostic Work-Up: Amyloid Biomarker Incongruences. J Alzheimers Dis 2021; 77:203-217. [PMID: 32716357 DOI: 10.3233/jad-200119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Discordance among amyloid biomarkers is a challenge to overcome in order to increase diagnostic accuracy in dementia. OBJECTIVES 1) To verify that cerebrospinal fluid (CSF) Aβ42/Aβ40 ratio (AβR) better agrees with Amyloid PET (Amy-PET) results compared to CSF Aβ42; 2) to detect differences among concordant positive, concordant negative, and discordant cases, basing the concordance definition on the agreement between CSF AβR and Amy-PET results; 3) to define the suspected underlying pathology of discordant cases using in vivo biomarkers. METHOD We retrospectively enrolled 39 cognitively impaired participants in which neuropsychological tests, apolipoprotein E genotype determination, TC/MRI, FDG-PET, Amy-PET, and CSF analysis had been performed. In all cases, CSF analysis was repeated using the automated Lumipulse method. In discordant cases, FDG-PET scans were evaluated visually and using automated classifiers. RESULTS CSF AβR better agreed with Amy-PET compared to CSF Aβ42 (Cohen's K 0.431 versus 0.05). Comparisons among groups did not show any difference in clinical characteristics except for age at symptoms onset that was higher in the 6 discordant cases with abnormal CSF AβR values and negative Amy-PET (CSF AβR+/AmyPET-). FDG-PET and all CSF markers (Aβ42, AβR, p-Tau, t-Tau) were suggestive of Alzheimer's disease (AD) in 5 of these 6 cases. CONCLUSION 1) CSF AβR is the CSF amyloid marker that shows the better level of agreement with Amy-PET results; 2) The use of FDG-PET and CSF-Tau markers in CSFAβR+/Amy-PET-discordant cases can support AD diagnosis; 3) Disagreement between positive CSF AβR and negative Amy-PET in symptomatic aged AD patients could be due to the variability in plaques conformation and a negative Amy-PET scan cannot be always sufficient to rule out AD.
Collapse
Affiliation(s)
- Gemma Lombardi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione Filippo Turati, Pistoia, Italy
| | | | | | - Cristina Polito
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Sonia Padiglioni
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | | | - Valentina Berti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Nuclear Medicine Unit, University of Florence, Florence, Italy
| | | | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione IRCCS Don Carlo Gnocchi, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,Fondazione IRCCS Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
50
|
Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simrén J, Montoliu‐Gaya L, Pannee J, O´Connor A, Weston PSJ, Lantero‐Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, Zetterberg H. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease. J Intern Med 2021; 290:583-601. [PMID: 34021943 PMCID: PMC8416781 DOI: 10.1111/joim.13332] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is increasingly prevalent worldwide, and disease-modifying treatments may soon be at hand; hence, now, more than ever, there is a need to develop techniques that allow earlier and more secure diagnosis. Current biomarker-based guidelines for AD diagnosis, which have replaced the historical symptom-based guidelines, rely heavily on neuroimaging and cerebrospinal fluid (CSF) sampling. While these have greatly improved the diagnostic accuracy of AD pathophysiology, they are less practical for application in primary care, population-based and epidemiological settings, or where resources are limited. In contrast, blood is a more accessible and cost-effective source of biomarkers in AD. In this review paper, using the recently proposed amyloid, tau and neurodegeneration [AT(N)] criteria as a framework towards a biological definition of AD, we discuss recent advances in biofluid-based biomarkers, with a particular emphasis on those with potential to be translated into blood-based biomarkers. We provide an overview of the research conducted both in CSF and in blood to draw conclusions on biomarkers that show promise. Given the evidence collated in this review, plasma neurofilament light chain (N) and phosphorylated tau (p-tau; T) show particular potential for translation into clinical practice. However, p-tau requires more comparisons to be conducted between its various epitopes before conclusions can be made as to which one most robustly differentiates AD from non-AD dementias. Plasma amyloid beta (A) would prove invaluable as an early screening modality, but it requires very precise tests and robust pre-analytical protocols.
Collapse
Affiliation(s)
- D. O. T. Alawode
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - A. J. Heslegrave
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - N. J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Old Age PsychiatryInstitute of Psychiatry, Psychology & NeuroscienceKing’s College LondonLondonUK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS FoundationLondonUK
| | - T. K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Simrén
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - L. Montoliu‐Gaya
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - J. Pannee
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - A. O´Connor
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - P. S. J. Weston
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. Lantero‐Rodriguez
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - A. Keshavan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - A. Snellman
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Turku PET CentreUniversity of TurkuTurkuFinland
| | - J. Gobom
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - R. W. Paterson
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - J. M. Schott
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - K. Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - N. C. Fox
- UK Dementia Research Institute at UCLLondonUK
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - H. Zetterberg
- From theDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| |
Collapse
|