1
|
Valletta M, Canevelli M, Gasparini F, Buscarnera S, Salzillo M, Triolo F, Calderón-Larrañaga A, Marengoni A, Vetrano DL, Grande G. Multimorbidity and fluid biomarkers of Alzheimer's disease: a systematic review. Eur Geriatr Med 2025:10.1007/s41999-025-01222-y. [PMID: 40392441 DOI: 10.1007/s41999-025-01222-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 04/16/2025] [Indexed: 05/22/2025]
Abstract
PURPOSE This systematic review aimed to summarize the evidence on the association between multimorbidity and fluid biomarkers of Alzheimer's disease (AD). METHODS We systematically searched PubMed, Web of Science, and Embase for studies investigating the association between multimorbidity-defined as the co-occurrence of multiple chronic conditions in the same individual-and levels of cerebrospinal fluid (CSF) or blood biomarkers of AD, focusing on the most established AD biomarkers (amyloid-beta, phosphorylated-tau, total-tau, neurofilament light chain, and glial fibrillary acidic protein). Studies were selected following PRISMA guidelines. RESULTS Out of 3,104 records, we identified 10 cross-sectional studies. Four studies assessed CSF biomarkers in dementia-free participants with mean age between 61.8 and 66.6 years, yielding mixed findings with no consistent association between multimorbidity and CSF biomarkers. Six studies focused on blood biomarkers in participants with mean age ranging from 66.5 to 76.4 years, five of which included individuals with dementia. Most of these studies reported an association between multimorbidity and elevated blood biomarker levels. CONCLUSIONS This review suggests a significant association between multimorbidity and AD blood biomarkers in older populations, while the results on CSF are mixed and inconsistent. Further research is needed, particularly longitudinal studies assessing both CSF and blood biomarkers within the same populations.
Collapse
Affiliation(s)
- Martina Valletta
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Marco Canevelli
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Department of Human Neuroscience, Sapienza University, Rome, Italy
- National Centre for Disease Prevention and Health Promotion, Italian National Institute of Health, Rome, Italy
| | - Francesca Gasparini
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Department of Medicine, Geriatrics Section, University of Padova, Padua, Italy
| | | | - Martina Salzillo
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Federico Triolo
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Amaia Calderón-Larrañaga
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Stockholm Gerontology Research Center, Stockholm, Sweden
| | - Alessandra Marengoni
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Davide Liborio Vetrano
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Stockholm Gerontology Research Center, Stockholm, Sweden
| | - Giulia Grande
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden.
- Stockholm Gerontology Research Center, Stockholm, Sweden.
| |
Collapse
|
2
|
Hazan J, Abel E, Rosa Grilo M, Alawode D, Laranjinha I, Heslegrave AJ, Liu KY, Schott JM, Howard R, Zetterberg H, Fox NC. How well do plasma Alzheimer's disease biomarkers reflect the CSF amyloid status? J Neurol Neurosurg Psychiatry 2025; 96:566-572. [PMID: 39694821 DOI: 10.1136/jnnp-2024-334122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Can plasma biomarkers as well as cerebrospinal fluid (CSF) perform in the separation of amyloid-beta-positive (Aβ+) vs amyloid-beta-negative (Aβ-) groups across an age range seen in an NHS cognitive disorder clinic? METHODS As part of the routine diagnostic investigation of 111 clinic patients who had contemporaneous blood and CSF samples taken, patients were categorised into Aβ+ and Aβ- groups based on their CSF in an Aβ42/40 ratio. We then evaluated four single molecule array (Simoa) Quanterix assays, quantifying single plasma analytes and ratios (p-tau217, p-tau217/Aβ42 ratio, p-tau181, p-tau181/Aβ42 ratio and Aβ42/40 ratio) in their ability to distinguish between these groups and the effect of age. RESULTS The median (range) age of participants was 66 (55-79) years with 48 females (43.2%). The areas under the curve (AUC), not accounting for age, for the ability to discriminate Aβ+ from Aβ- groups were plasma p-tau217 AUC=0.94, Aβ42/40 AUC=0.78 and p-tau181 AUC=0.77. Combining p-tau217/Aβ42 increased the AUC to 0.97. The difference between the groups was influenced by age with less separation in older individuals: a significant negative interaction term between age and group for plasma p-tau217 concentrations (-0.037, p=0.013) and p-tau217/Aβ42 ratio (-0.007, p=0.008). CONCLUSIONS There was variable performance of plasma biomarkers to recapitulate the CSF assay. Both p-tau217 and p-tau217/Aβ42 showed excellent promise as surrogates of CSF amyloid status, although with slightly reduced performance in older individuals. There was poorer discriminatory ability for p-tau181 and Aβ42/40. Further research is needed to address potential age-related confounds.
Collapse
Affiliation(s)
| | - Emily Abel
- UCL, UK Dementia Research Institute, London, UK
| | | | | | | | | | | | | | | | - Henrik Zetterberg
- UCL, UK Dementia Research Institute, London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, Goteborg, Sweden
| | - Nick C Fox
- UCL, UK Dementia Research Institute, London, UK
- UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
3
|
Álvarez-Sánchez L, Peña-Bautista C, Ferré-González L, Balaguer Á, Amengual JL, Baquero M, Cháfer-Pericás C. Promising clinical tools for specific Alzheimer disease diagnosis from plasma pTau217 and ApoE genotype in a cognitive disorder unit. Sci Rep 2025; 15:16316. [PMID: 40348819 PMCID: PMC12065895 DOI: 10.1038/s41598-025-01511-3] [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: 12/24/2024] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on cerebrospinal fluid (CSF) biomarkers or amyloid PET. Alternatives for AD diagnosis from blood samples are needed to develop a fully-automated early-diagnosis approach, potentially implemented in a cognitive disorder unit. Plasma p-Tau217 was determined in patients diagnosed with AD (n = 134) or non-AD (n = 132), from CSF biomarkers (Aβ42/Aβ40). A logistic regression model was developed. The predictive performance was assessed using a training set (70% of data) and internally validated with a test set (30% of data) and 1000 iterations. A nomogram and a double cut-off strategy were proposed to visualize the model results, and stratify patients (AD, non-AD, uncertain), respectively. The model (plasma p-Tau217, ApoE, age) showed satisfactory performance (AUC 0.94, sensitivity 0.85, specificity 0.89); so, together with the corresponding nomogram, it could be applied in specialized clinical contexts. The model including only plasma p-Tau217 (AUC 0.93, sensitivity 0.72, specificity 0.96) would be a useful approach in less specialized clinics. The corresponding two-cut-off strategy for the first model were as AD probability (< 0.41 non-AD, > 0.57 AD). This study provided clinical tools (nomogram, double cut-off) for identifying Aβ positivity at a cognitive disorder unit, which would lead to reduce the CSF analysis.
Collapse
Affiliation(s)
- Lourdes Álvarez-Sánchez
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Carmen Peña-Bautista
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Laura Ferré-González
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Ángel Balaguer
- Plataforma de Big Data, IA y Bioestadística, Instituto de Investigación Sanitaria La Fe, 46026, Valencia, Spain
| | - Julián Luis Amengual
- Plataforma de Big Data, IA y Bioestadística, Instituto de Investigación Sanitaria La Fe, 46026, Valencia, Spain
| | - Miguel Baquero
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
- Division of Neurology, Instituto de Investigación Sanitaria La Fe, 46026, Valencia, Spain
| | - Consuelo Cháfer-Pericás
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| |
Collapse
|
4
|
Ossenkoppele R, Salvadó G, Janelidze S, Pichet Binette A, Bali D, Karlsson L, Palmqvist S, Mattsson-Carlgren N, Stomrud E, Therriault J, Rahmouni N, Rosa-Neto P, Coomans EM, van de Giessen E, van der Flier WM, Teunissen CE, Jonaitis EM, Johnson SC, Villeneuve S, Benzinger TLS, Schindler SE, Bateman RJ, Doecke JD, Doré V, Feizpour A, Masters CL, Rowe C, Wiste HJ, Petersen RC, Jack CR, Hansson O. Plasma p-tau217 and tau-PET predict future cognitive decline among cognitively unimpaired individuals: implications for clinical trials. NATURE AGING 2025; 5:883-896. [PMID: 40155777 DOI: 10.1038/s43587-025-00835-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/12/2025] [Indexed: 04/01/2025]
Abstract
Plasma p-tau217 and tau positron emission tomography (PET) are strong prognostic biomarkers in Alzheimer's disease (AD), but their relative performance in predicting future cognitive decline among cognitively unimpaired (CU) individuals is unclear. In a head-to-head comparison study including nine cohorts and 1,474 individuals, we show that plasma p-tau217 and medial temporal lobe tau-PET signal display similar associations with cognitive decline on a global cognitive composite test (R2PET = 0.34 versus R2plasma = 0.33, Pdifference = 0.653) and with progression to mild cognitive impairment (hazard ratio (HR)PET = 1.61 (1.48-1.76) versus HRplasma = 1.57 (1.43-1.72), Pdifference = 0.322). Combined plasma and PET models were superior to the single-biomarker models (R2 = 0.35, P < 0.01). Sequential selection using plasma phosphorylated tau at threonine 217 (p-tau217) and then tau-PET reduced the number of participants required for a clinical trial by 94%, compared to a 76% reduction when using plasma p-tau217 alone. Thus, plasma p-tau217 and tau-PET showed similar performance for predicting future cognitive decline in CU individuals, and their sequential use enhances screening efficiency for preclinical AD trials.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Emma M Coomans
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Elsmarieke van de Giessen
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sylvia Villeneuve
- Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - James D Doecke
- Australian eHealth Research Centre, Commonwealth Scientific and Industrial Research Organization, Melbourne, Victoria, Australia
| | - Vincent Doré
- Australian eHealth Research Centre, Commonwealth Scientific and Industrial Research Organization, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - Azadeh Feizpour
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Christopher Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
5
|
Ashton NJ, Benedet AL, Molfetta GD, Pola I, Anastasi F, Fernández‐Lebrero A, Puig‐Pijoan A, Keshavan A, Schott J, Tan K, Simrén J, Gomes BF, Montoliu‐Gaya L, Isaacson R, Bongianni M, Tolassi C, Cantoni V, Alberici A, Padovani A, Zanusso G, Pilotto A, Borroni B, Suárez‐Calvet M, Blennow K, Zetterberg H. Biomarker discovery in Alzheimer's and neurodegenerative diseases using Nucleic Acid Linked Immuno-Sandwich Assay. Alzheimers Dement 2025; 21:e14621. [PMID: 40401628 PMCID: PMC12096316 DOI: 10.1002/alz.14621] [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: 08/05/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 05/23/2025]
Abstract
INTRODUCTION Recent advancements in immunological methods accurately quantify biofluid biomarkers for Alzheimer's disease (AD) pathology. Despite progress, more biomarkers, ideally in blood, are needed for effective disease monitoring for AD and other neurodegenerative proteinopathies. METHODS We used the Nucleic Acid Linked Immuno-Sandwich Assay (NULISA) central nervous system panel for biomarker quantification in plasma, serum, and cerebrospinal fluid of patients with AD, mild cognitive impairment, Lewy body dementia, progranulin (GRN) mutation carriers. RESULTS NULISA identified phosphorylated tau217 and neurofilament light chain as the most deregulated biomarkers in the AD continuum and GRN mutation carriers, respectively. Importantly, numerous novel proteomic changes were observed in each disease endophenotype, which included synaptic processing, inflammation, microglial reactivity, TAR DNA-binding protein 43, and α-synuclein pathology. DISCUSSION We underline the potential of next-generation biomarker identification tools to detect novel proteomic features that also incorporate established biomarkers. These findings highlight the importance of continued biomarker discovery to improve treatment decisions and help us better understand the complexities of neurodegenerative disorders. HIGHLIGHTS The, direct, or indirect, measures in blood that complement phosphorylated tau (p-tau)217 for other proteinopathies or disease progression are urgently needed. Significant novel proteomic changes were observed in each disease endophenotype in plasma, serum, and cerebrospinal fluid, which included proteins involved in synaptic processing, inflammation, microglial reactivity, TAR DNA-binding protein 43, and α-synuclein pathology. Nucleic Acid Linked Immuno-Sandwich Assay continued to unbiasely highlight p-tau217 and neurofilament light chain as the most significantly deregulated blood biomarkers in the Alzheimer's disease continuum and progranulin mutation carriers, respectively.
Collapse
Affiliation(s)
- Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Banner Alzheimer's Institute and University of ArizonaPhoenixArizonaUSA
- Banner Sun Health Research InstituteSun CityArizonaUSA
| | - Andrea L. Benedet
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Guglielmo Di Molfetta
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Ilaria Pola
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Federica Anastasi
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Aida Fernández‐Lebrero
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Albert Puig‐Pijoan
- Hospital del Mar Research InstituteBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
- Department of MedicineUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Ashvini Keshavan
- Dementia Research CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Jonathan Schott
- Dementia Research CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Kubra Tan
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Joel Simrén
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Bárbara Fernandes Gomes
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Laia Montoliu‐Gaya
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Richard Isaacson
- Department of NeurologyWeill Cornell Medicine and New York PresbyterianNew YorkNew YorkUSA
- Department of NeurologyFlorida Atlantic University, Charles E. Schmidt College of MedicineBoca RatonFloridaUSA
| | - Matilde Bongianni
- Department of Neurosciences, Biomedicine, and Movement SciencesPoliclinico G. B. Rossi, University of VeronaVeronaItaly
| | - Chiara Tolassi
- Clinical Investigation in LaboratoryMaggiore Hospital ASST‐CremaCremaItaly
| | - Valentina Cantoni
- Department of Clinical and Experimental Sciences, Neurology UnitUniversity of BresciaBresciaItaly
| | - Antonella Alberici
- Department of Clinical and Experimental Sciences, Neurology UnitUniversity of BresciaBresciaItaly
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology UnitUniversity of BresciaBresciaItaly
- Department of Continuity of Care and FrailtyAzienda Socio Sanitaria Territoriale (ASST) Spedali CiviliBresciaItaly
- Laboratory of Digital Neurology and BiosensorsUniversity of BresciaBresciaItaly
- Brain Health CenterUniversity of BresciaBresciaItaly
| | - Gianluigi Zanusso
- Department of Neuroscience, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Andrea Pilotto
- Department of Clinical and Experimental Sciences, Neurology UnitUniversity of BresciaBresciaItaly
- Department of Continuity of Care and FrailtyAzienda Socio Sanitaria Territoriale (ASST) Spedali CiviliBresciaItaly
- Laboratory of Digital Neurology and BiosensorsUniversity of BresciaBresciaItaly
- Brain Health CenterUniversity of BresciaBresciaItaly
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, Neurology UnitUniversity of BresciaBresciaItaly
- Department of Continuity of Care and FrailtyAzienda Socio Sanitaria Territoriale (ASST) Spedali CiviliBresciaItaly
| | - Marc Suárez‐Calvet
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain InstituteICM, Pitié‐Salpêtrière Hospital, Sorbonne University, Hôpital PitiéParisFrance
- 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 USTCHefeiP. R. China
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Dementia Research CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public Health, University of Wisconsin–MadisonMadisonWisconsinUSA
| |
Collapse
|
6
|
Noguchi‐Shinohara M, Shuta K, Murakami H, Mori Y, Komatsu J, Kobayashi C, Hersch S, Horie K, Ono K. Lecanemab-Associated Amyloid-β Protofibril in Cerebrospinal Fluid Correlates with Biomarkers of Neurodegeneration in Alzheimer's Disease. Ann Neurol 2025; 97:993-1006. [PMID: 39761671 PMCID: PMC12010060 DOI: 10.1002/ana.27175] [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: 08/14/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 04/22/2025]
Abstract
OBJECTIVE The Clarity AD phase III trial showed that lecanemab reduced amyloid markers in early Alzheimer's disease (AD) and resulted in less decline on measures of cognition and function than placebo. Herein, we aimed to characterize amyloid-β (Aβ) protofibril (PF) captured by lecanemab in human cerebrospinal fluid (CSF) from living participants with different stages in AD, which enable an enhanced understanding of the dynamic changes of lecanemab-associated Aβ-PF (Lec-PF) in vivo. METHODS We newly developed a unique and highly sensitive immunoassay method using lecanemab that selectively captures Lec-PF. The CSF level of Lec-PF, Aβ42, Aβ40, p-tau181, p-tau 217, total tau, and neurogranin were measured in Japanese participants (n = 163). The participants in this study consisted of 48 cognitively unimpaired Aβ-negative (CU-), 8 cognitively impaired diagnosed as suspected non-Alzheimer's disease pathophysiology, 9 cognitively unimpaired Aβ-positive (CU+), 34 Aβ-positive with mild cognitive impairment (MCI+), and 64 Aβ-positive with AD dementia (AD+). RESULTS The CSF Lec-PF levels significantly increased in the groups of MCI+ and AD+ compared with CU- group. Notably, CSF Lec-PF showed modest correlation with plaque-associated biomarkers in Aβ-positive participants and stronger correlation with neurodegeneration biomarkers, such as CSF total tau and neurogranin, suggesting that CSF Lec-PF levels proximally reflect neurodegeneration as well as the amount of senile amyloid plaques. INTERPRETATION This is the first report describing Aβ-PF species captured by lecanemab in human CSF and supporting that Lec-PF is correlated with neurodegeneration in AD and may explain the mechanism of the clinical effect of lecanemab. ANN NEUROL 2025;97:993-1006.
Collapse
Affiliation(s)
| | - Kazuyoshi Shuta
- Deep Human Biology Learning (DHBL)Eisai Co., LtdTsukubaJapan
| | - Hidetomo Murakami
- Department of Neurology, School of MedicineShowa UniversityTokyoJapan
| | - Yukiko Mori
- Department of Neurology, School of MedicineShowa UniversityTokyoJapan
| | - Junji Komatsu
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | | | - Steven Hersch
- Deep Human Biology Learning (DHBL)Eisai Inc.NutleyNJUSA
| | - Kanta Horie
- Deep Human Biology Learning (DHBL)Eisai Inc.NutleyNJUSA
| | - Kenjiro Ono
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| |
Collapse
|
7
|
Bathla S, Datta D, Bolat D, Woo E, Duque A, Arellano JI, Arnsten AFT, Nairn AC. Dysregulated calcium signaling in the aged macaque entorhinal cortex associated with tau hyperphosphorylation. Front Aging Neurosci 2025; 17:1549770. [PMID: 40365352 PMCID: PMC12069431 DOI: 10.3389/fnagi.2025.1549770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Tau pathology in sporadic Alzheimer's disease (AD) follows a distinct pattern, beginning in the entorhinal cortex (ERC) and spreading to interconnected brain regions. Early-stage tau pathology, characterized by soluble phosphorylated tau, is difficult to study in human brains post-mortem due to rapid dephosphorylation. Methods Rhesus macaques, which naturally develop age-related tau pathology resembling human AD, provide an ideal model for investigating early tau etiology. This study examines the molecular processes underlying tau pathology in the macaque ERC, focusing on calcium and inflammatory signaling pathways using biochemical and immunohistochemistry. Results Our findings reveal an age-related decrease in PDE4 phosphodiesterase that hydrolyzes cAMP and increases in calpain-2 and glutamate carboxypeptidase II that occur in parallel with early-stage tau hyperphosphorylation at multiple epitopes (pS214-tau, pT181-tau, pT217-tau). Discussion These findings suggest that dysregulated calcium signaling in ERC, beginning in middle-age, may prime tau for hyperphosphorylation, potentially driving the early stages of AD, advancing our understanding of how ERC vulnerabilities contribute to neurodegeneration in AD.
Collapse
Affiliation(s)
- Shveta Bathla
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dinara Bolat
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Elizabeth Woo
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Jon I. Arellano
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Amy F. T. Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Angus C. Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
8
|
Vanderlinden G, Vandenberghe R, Vandenbulcke M, Van Laere K. The Current Role of Tau PET Imaging in Neurodegeneration. Semin Nucl Med 2025:S0001-2998(25)00031-5. [PMID: 40263023 DOI: 10.1053/j.semnuclmed.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Neurodegenerative tauopathies are characterized by the pathological hyperphosphorylation of tau proteins that subsequently form aggregates. Tau PET tracers with affinity to bind these pathological tau aggregates have been developed to measure disease progression and to support therapeutic drug development. In this review, we summarize the pathophysiology of tau throughout the range of neurodegenerative tauopathies. We outline the available first- and second-generation tau PET tracers, with a focus on new tau PET tracer developments, and discuss the quantification of tau PET images. Next, we summarize how tau PET relates to cerebrospinal fluid and plasma tau biomarkers. Finally, we review the current recommendations on the clinical use of tau PET versus fluid tau biomarkers in diagnosis, prognosis and treatment development.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Research Group Psychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Division of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Pola I, Ashton NJ, Antônio De Bastiani M, Brum WS, Rahmouni N, Tan K, Machado LS, Servaes S, Stevenson J, Tissot C, Therriault J, Pascoal TA, Blennow K, Zetterberg H, Zimmer ER, Rosa‐Neto P, Benedet AL. Exploring inflammation-related protein expression and its relationship with TSPO PET in Alzheimer's disease. Alzheimers Dement 2025; 21:e70171. [PMID: 40289873 PMCID: PMC12035552 DOI: 10.1002/alz.70171] [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: 11/06/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/30/2025]
Abstract
INTRODUCTION To understand the role of neuroinflammation in Alzheimer's disease (AD), we characterized immune-related proteins in central and peripheral biofluids. METHODS Selection of participants from the Translational Biomarker of Aging and Dementia (TRIAD) cohort with available translocator protein (TSPO) positron emission tomography (PET), cerebrospinal fluid (CSF) (n = 97), and plasma (n = 165). Biofluid samples analyzed with Olink technology (368 inflammation proteins). RESULTS Elevated proteins levels in CSF of TSPO-positive individuals were identified. Functional enrichment analysis of CSF proteins revealed processes implicated in AD (MAPK, ERK cascades, cytokine, and leukocyte signaling). Selected candidates (CXCL1 and TNFRSF11B) showed high correlation with each other in CSF and with TSPO PET signal, but weaker associations with amyloid and tau PET. No significantly changed proteins in plasma between TSPO groups were found. DISCUSSION This explorative study identified two potential targets in CSF showing correlations with TSPO, amyloid and tau PET, suggesting a direct link between neuroinflammation, expression of these proteins and their potential implication in AD. HIGHLIGHTS Several proteins are elevated in CSF of TSPO PET-positive individuals, linking them to neuroinflammation. Elevated CSF proteins were enriched in pathways such as MAPK, ERK, and cytokine signaling, linking them to the AD pathophysiology. Candidate proteins (CXCL1 and TNFRSF11B) correlated strongly with TSPO PET, particularly in brain regions known to be affected in AD. Although none of the plasma proteins remained significant after multiple comparisons correction when comparing their expression between TSPO groups, as done for CSF, candidate CSF proteins were found to correlate with plasmatic proteins, highlighting the complexity of the immune system.
Collapse
Affiliation(s)
- Ilaria Pola
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Banner Alzheimer's Institute and University of ArizonaPhoenixArizonaUSA
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
- King's College LondonInstitute of PsychiatryPsychology & NeuroscienceMaurice Wohl Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
| | - Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: BiochemistryUniversidade Federal do Rio Grande do Sul (UFRGS)Porto AlegreBrazil
| | - Wagner S. Brum
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Graduate Program in Biological Sciences: BiochemistryUniversidade Federal do Rio Grande do Sul (UFRGS)Porto AlegreBrazil
| | - Nesrine Rahmouni
- McGill Centre for Studies in AgingMcGill UniversityVerdunQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityVerdunQuebecCanada
| | - Kubra Tan
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
| | - Luiza Santos Machado
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
| | - Stijn Servaes
- McGill Centre for Studies in AgingMcGill UniversityVerdunQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityVerdunQuebecCanada
| | - Jenna Stevenson
- McGill Centre for Studies in AgingMcGill UniversityVerdunQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityVerdunQuebecCanada
| | - Cécile Tissot
- McGill Centre for Studies in AgingMcGill UniversityVerdunQuebecCanada
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Joseph Therriault
- McGill Centre for Studies in AgingMcGill UniversityVerdunQuebecCanada
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Tharick A. Pascoal
- Departments of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: BiochemistryUniversidade Federal do Rio Grande do Sul (UFRGS)Porto AlegreBrazil
- McGill Centre for Studies in AgingMcGill UniversityVerdunQuebecCanada
- Department of PharmacologyGraduate Program in Biological Sciences: Pharmacology and TherapeuticsUniversidade Federal do Rio Grande do Sul (UFRGS)Porto AlegreBrazil
- Brain Institute of Rio Grande do SulPUCRSPorto AlegreBrazil
| | - Pedro Rosa‐Neto
- McGill Centre for Studies in AgingMcGill UniversityVerdunQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityVerdunQuebecCanada
| | - Andréa L. Benedet
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
| |
Collapse
|
10
|
Wei M, Zhang Y, Wang M, Zhong J, Chai W, Hu W, Tao F, Wang L, Zhang Q, Yu X, Shi R, Li C, Song Z, Chen G, Zhang S, Jiang J, Han Y, for the Alzheimer's Disease Neuroimaging Initiative. Plasma biomarkers as core mediators in functional network abnormalities: Evidence from a two-cohort study. Alzheimers Dement 2025; 21:e70114. [PMID: 40329658 PMCID: PMC12056302 DOI: 10.1002/alz.70114] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/11/2025] [Accepted: 02/26/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION Researchers have reported an association among amyloid beta (Aβ), tau deposition, and functional networks. Nevertheless, whether plasma biomarkers mediate this process remains unclear. METHODS Three hundred and forty-eight participants with available plasma biomarkers, Aβ positron emission tomography (PET), and resting-state functional magnetic resonance imaging were obtained from two independent cohorts: SILCODE (n = 147) and ADNI (n = 201). Correlations among plasma biomarkers, functional connectivity, and global standardized uptake value ratio (SUVR) were assessed, and mediating effects were analyzed to explore underlying pathways. RESULTS Plasma biomarkers (p-tau181, p-tau217, neurofilament light, glial fibrillary acidic protein, Aβ42/40) demonstrated significant correlations with global SUVR and functional connectivity across networks (p < 0.05). Significant functional connectivity variations in different networks were observed across various Aβ stages, with differences mediated by plasma biomarkers. The crucial pathway exhibited fully mediated effects: Aβ PET SUVR-plasma biomarkers (mainly p-tau181 and p-tau217) - various functional networks. DISCUSSION Our study highlights the core mediating role of plasma biomarkers (mainly p-tau181 and p-tau217) in the progression of Aβ accumulation and in various functional network alterations. HIGHLIGHTS Plasma biomarkers demonstrated significant mediating effects on Aβ deposition and functional network alterations across different Aβ stages in both East Asian and Western cohorts. Significant functional connectivity variations in different brain networks were observed throughout AD progression in two cohorts, particularly across various Aβ stages. Among all plasma biomarkers, p-tau217 and p-tau181 demonstrated more comprehensive and fully mediating effects compared to other biomarkers, influencing connectivity alterations in the various functional networks. The plasma biomarkers exhibited great potential for tracking pathologic progression, functional network alterations, and early disease identification in East Asian and Western cohorts.
Collapse
Affiliation(s)
- Min Wei
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Ying Zhang
- Institute of Biomedical Engineering, School of MedicineShanghai UniversityShanghaiChina
| | - Min Wang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Jiayi Zhong
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Wenhui Chai
- Department of GeriatricsXinjiang Changji People's HospitalXinjiangChina
| | - Wenjing Hu
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Fengli Tao
- The Central Hospital of KaramayXinjiangChina
| | - Luyao Wang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Qi Zhang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Xianfeng Yu
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- Department of NeurologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Rong Shi
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Chenyang Li
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Ziyan Song
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Guanqun Chen
- Department of NeurologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Shuyu Zhang
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- The Central Hospital of KaramayXinjiangChina
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical EngineeringHainan UniversitySanyaChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
| | | |
Collapse
|
11
|
Lan G, Li A, Gonzalez-Ortiz F, Lv J, Ran W, Cai Y, Sun P, Liu L, Yang J, Zhang L, He Z, Fang L, Zhou X, Zhu Y, Liu Z, Fan X, Chen X, Xu L, Wang Q, Wang X, Sun K, Cheng G, Han Y, Blennow K, Wang L, Ran P, Guo T. Higher plasma soluble TREM2 correlates with reduced cerebral tau accumulation in Alzheimer's disease. Mol Psychiatry 2025:10.1038/s41380-025-02976-4. [PMID: 40169800 DOI: 10.1038/s41380-025-02976-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
Loss-of-function mutation of triggering receptor expressed on myeloid cell 2 (TREM2) is associated with increased risks for Alzheimer's disease (AD). Recent animal studies reveal that the activation of peripheral TREM2 signaling may affect cerebral β-amyloid (Aβ) and tau aggregates. However, the underlying relationship between peripheral TREM2 and brain AD pathology has not yet been well-elucidated in the aging population. In this study, we collected 318 Chinese older adults with Aβ PET and plasma biomarker measures, including soluble TREM2 (sTREM2) and glial fibrillary acidic protein (GFAP), a proxy for astrocyte reactivity. Additionally, 129 participants underwent tau PET scans. We explored the association between plasma sTREM2, GFAP, and primary AD pathology. Plasma sTREM2 was negatively associated with reduced temporal tau PET burden in participants with abnormal Aβ and tau pathology. Higher plasma sTREM2 was related to the weaker association of Aβ pathology and plasma phosphorylated tau with tau PET increases. In contrast, elevated plasma GFAP was related to greater Aβ and tau PET burden along with stronger Aβ-related tau accumulation. Finally, higher plasma sTREM2 was linked to attenuated strength of the association between plasma GFAP and tau PET increases at both pre-defined regions of interest and voxel levels. Altogether, our findings suggest distinct relationships between plasma sTREM2 and GFAP with cerebral tau pathology, providing novel insights into the roles of peripheral TREM2 signaling and astrocytic reactivity in AD neuropathological development. This study has important implications, such as targeting the peripheral TREM2 signature, which may be a potential strategy for future AD therapies.
Collapse
Affiliation(s)
- Guoyu Lan
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Anqi Li
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region, China
| | - Fernando Gonzalez-Ortiz
- Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, 40530, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 41345, Sweden
| | - Jieqin Lv
- Department of Nuclear Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510641, China
| | - Wenqing Ran
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yue Cai
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Pan Sun
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518132, China
| | - Lin Liu
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518132, China
| | - Jie Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Laihong Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Zhengbo He
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Lili Fang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xin Zhou
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yalin Zhu
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region, China
| | - Zhen Liu
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xiang Fan
- Department of Medical Imaging, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Linsen Xu
- Department of Medical Imaging, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, China
| | - Qingyong Wang
- Department of Neurology, Shenzhen Guangming District People's Hospital, Shenzhen, 518107, China
| | - Xinlu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510641, China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Guanxun Cheng
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China
| | - Ying Han
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China
- National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
- The Central Hospital of Karamay, Karamay, Xinjiang, 834000, China
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, 75646, France
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, 40530, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 41345, Sweden
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, 230000, China
- Huaqiao Hospital, Jinan University, Guangzhou, 510641, China
| | - Lu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Pengcheng Ran
- Department of Nuclear Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510641, China.
| | - Tengfei Guo
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
- Peking University Shenzhen Graduate School, Peking University, Shenzhen, 518132, China.
| |
Collapse
|
12
|
Horie K, Salvadó G, Koppisetti RK, Janelidze S, Barthélemy NR, He Y, Sato C, Gordon BA, Jiang H, Benzinger TLS, Stomrud E, Holtzman DM, Mattsson-Carlgren N, Morris JC, Palmqvist S, Ossenkoppele R, Schindler SE, Hansson O, Bateman RJ. Plasma MTBR-tau243 biomarker identifies tau tangle pathology in Alzheimer's disease. Nat Med 2025:10.1038/s41591-025-03617-7. [PMID: 40164726 DOI: 10.1038/s41591-025-03617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
Insoluble tau aggregates within neurofibrillary tangles are a defining neuropathological feature of Alzheimer's disease (AD) and closely correlate with clinical symptoms. Although tau pathology can be assessed using tau positron emission tomography, a more accessible biomarker is needed for diagnosis, prognosis and tracking treatment effects. Here we present a new plasma tau species, the endogenously cleaved, microtubule-binding region containing residue 243 (eMTBR-tau243), which specifically reflects tau tangle pathology. Across the AD spectrum in three different cohorts (n = 108, 55 and 739), plasma eMTBR-tau243 levels were significantly elevated at the mild cognitive impairment stage and increased further in dementia. Plasma eMTBR-tau243 showed strong associations with tau positron emission tomography binding (β = 0.72, R2 = 0.56) and cognitive performance (β = 0.60, R2 = 0.40), outperforming other plasma tau (%p-tau217 and %p-tau205) biomarkers. These results suggest that plasma eMTBR-tau243 may be useful for estimating the tauopathy load in AD, thereby improving the diagnostic evaluation of AD in clinical practice and monitoring the efficacy of tau-targeted therapies in clinical trials.
Collapse
Affiliation(s)
- Kanta Horie
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Eisai Inc., Nutley, NJ, USA.
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rama K Koppisetti
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Nicolas R Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chihiro Sato
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| | - Randall J Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
13
|
Martínez-Dubarbie F, Guerra-Ruiz A, López-García S, Lage C, Fernández-Matarrubia M, Nevado-Cáceres Á, Rivera-Sánchez M, Valera-Barrero A, Pozueta-Cantudo A, García-Martínez M, Corrales-Pardo A, Bravo M, López-Hoyos M, Irure-Ventura J, de Lucas EM, Drake-Pérez M, Cahuana-Santamaría NH, García-Unzueta MT, Sánchez-Juan P, Rodríguez-Rodríguez E. Diagnostic performance of plasma p-tau217 in a memory clinic cohort using the Lumipulse automated platform. Alzheimers Res Ther 2025; 17:68. [PMID: 40140924 PMCID: PMC11948696 DOI: 10.1186/s13195-025-01719-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Plasma biomarkers for Alzheimer's disease (AD) are a promising tool for accessible and accurate biological diagnostics. However, data in clinical practice are needed to better understand their diagnostic and prognostic ability in memory unit patients. METHODS We analyzed plasma phosphorylated tau at threonine 217 (p-tau217) and neuroflament light chain (NfL) levels and AD cerebrospinal fluid (CSF) biomarkers in a group of 493 subjects using the Lumipulse G600II platform. The sample includes 340 patients from our memory unit (142 dementia, 186 mild cognitive impairment, and 12 with subjective complaints) and 153 cognitively unimpaired volunteers. We have correlated plasma and CSF biomarkers; we have analyzed plasma biomarker levels as a function of clinical diagnosis, cognitive status and amyloid status. We have also studied the ability of p-tau217 to discriminate between amyloid-positive and -negative subjects according to CSF using receiver operating characteristic curves. RESULTS Plasma p-tau217 correlated significantly with CSF Aβ42/Aβ40 (Rho = -0.75; p-value < 0.001), p-tau181 (r = 0.66; p-value < 0.001), and t-tau (r = 0.59; p-value < 0.001). Plasma NfL correlated with CSF NfL (r = 0.48; p-value < 0.001). By clinical diagnosis, plasma p-tau217 levels showed to be higher in AD patients than in healthy controls (difference = 0.63 pg/ml; p-value < 0.001), FTD (difference = 0.60 pg/ml; p-value < 0.001), and nondegenerative dementias (difference = 0.61 pg/ml; p-value < 0.001). Plasma p-tau217 showed an area under the curve of 0.95 to discriminate between A + and A- subjects (95%CI 0.93-0.97). CONCLUSION Plasma p-tau217 shows excellent results for detecting amyloid pathology at brain level in a clinical setting with an AUC of 0.95. It is a highly specific marker of AD and increases progressively along the disease continuum. Using plasma p-tau217 as an initial diagnostic tool with cut-offs at sensitivities and specificities of 95 or 97.5% could save between 57.4-84.8% of LP/PETs with diagnostic accuracies of 95-97%. Plasma NfL increases progressively at different cognitive stages.
Collapse
Affiliation(s)
- Francisco Martínez-Dubarbie
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain.
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain.
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28220, Spain.
| | - Armando Guerra-Ruiz
- Biochemistry and Clinical Analysis Department, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
| | - Sara López-García
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28220, Spain
| | - Carmen Lage
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28220, Spain
- Atlantic Fellow for Equity in Brain health, Global Brain Health Institute, University of California, San Francisco, 94158, USA
| | - Marta Fernández-Matarrubia
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28220, Spain
| | - Álvaro Nevado-Cáceres
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
| | - María Rivera-Sánchez
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - Andrea Valera-Barrero
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
| | - Ana Pozueta-Cantudo
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - María García-Martínez
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - Andrea Corrales-Pardo
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - María Bravo
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28220, Spain
| | - Marcos López-Hoyos
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Immunology Department, Marqués de Valdecilla University Hospital, Santander, 39008, Spain
- Molecular Biology Department, University of Cantabria, Santander, 39011, Spain
| | - Juan Irure-Ventura
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Immunology Department, Marqués de Valdecilla University Hospital, Santander, 39008, Spain
| | - Enrique Marco de Lucas
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Radiology Department, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Marta Drake-Pérez
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Radiology Department, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Nancy Heidy Cahuana-Santamaría
- Biochemistry and Clinical Analysis Department, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
| | - María Teresa García-Unzueta
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Biochemistry and Clinical Analysis Department, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
| | - Pascual Sánchez-Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28220, Spain
- Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, Madrid, 28031, Spain
| | - Eloy Rodríguez-Rodríguez
- Neurology Service, Marqués de Valdecilla University Hospital, Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28220, Spain
- Medicine and Psychiatry Department, University of Cantabria, Santander, 39011, Spain
| |
Collapse
|
14
|
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] [Download PDF] [Figures] [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
|
15
|
Lantero-Rodriguez J, Montoliu-Gaya L, Ashton NJ, Pola I, Therriault J, Rahmouni N, Brum WS, Servaes S, Stevenson J, Di Molfetta G, Arslan B, Klostranec J, Vitali P, Montembeault M, Gauthier S, Tissot C, Macedo AC, Pascoal TA, Jeromin A, Gobom J, Blennow K, Zetterberg H, Rosa-Neto P, Benedet AL. Biofluid-based staging of Alzheimer's disease. Acta Neuropathol 2025; 149:27. [PMID: 40095069 PMCID: PMC11913990 DOI: 10.1007/s00401-025-02863-w] [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: 11/06/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Recently, conceptual systems for the in vivo staging of Alzheimer's disease (AD) using fluid biomarkers have been suggested. Thus, it is important to assess whether available fluid biomarkers can successfully stage AD into clinically and biologically relevant categories. In the TRIAD cohort, we explored whether p-tau217, p-tau205 and NTA-tau (biomarkers of early, intermediate and late AD pathology, respectively) have potential for biofluid-based staging in cerebrospinal fluid (CSF; n = 219) and plasma (n = 150), and compared them in a paired CSF and plasma subset (n = 76). Our findings suggest a good concordance between biofluid staging and underlying pathology when classifying amyloid-positivity into three categories based on neurofibrillary pathology: minimal/non-existent (p-tau217 positive), early-to-intermediate (p-tau217 and p-tau205 positivity), and advanced tau tangle deposition (p-tau217, p-tau205 and NTA-tau positive), as indexed by tau-PET. Discordant cases accounted for 4.6% and 13.3% of all CSF and plasma measurements respectively (9.2% and 11.8% in paired samples). Notably, CSF- and plasma-based staging matched one another in 61.7% of the cases, while approximately 32% of the remaining participants were one to three biofluid stages higher in CSF as compared to plasma. Overall, these exploratory results suggest that biofluid staging of AD holds potential for offering valuable insights into underlying AD hallmarks and disease severity. However, its applicability beyond molecular characterization at research settings has yet to be demonstrated.
Collapse
Grants
- Members of the CIHR-CCNA Canadian Consortium of Neurodegeneration in Aging. Colin J. Adair Charitable Foundation.
- Swedish Research Council (#2017-00915 and #2022-00732)
- Swedish Alzheimer Foundation (#AF-930351, #AF-939721 and #AF-968270)
- Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006)
- Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240)
- European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236)
- Alzheimer’s Association 2021 Zenith Award (ZEN-21-848495)
- Alzheimer’s Association 2022-2025 Grant (SG-23-1038904 QC)
- Swedish Research Council (#2023-00356; #2022-01018 and #2019-02397)
- European Union’s Horizon Europe research and innovation programme under grant agreement No 101053962
- Swedish State Support for Clinical Research (#ALFGBG-71320)
- Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862)
- AD Strategic Fund and the Alzheimer's Association (#ADSF-21-831376-C, #ADSF-21-831381-C, #ADSF-21-831377-C, and #ADSF-24-1284328-C)
- European Partnership on Metrology, co-financed from the European Union’s Horizon Europe Research and Innovation Programme and by the Participating States (NEuroBioStand, #22HLT07)
- Bluefield Project
- Cure Alzheimer’s Fund
- Olav Thon Foundation
- Erling-Persson Family Foundation
- Familjen Rönströms Stiftelse
- Stiftelsen för Gamla Tjänarinnor
- Hjärnfonden, Sweden (#FO2022-0270)
- European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE)
- European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694)
- National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
- UK Dementia Research Institute at UCL (UKDRI-1003)
- Weston Brain Institute, Canadian Institutes of Health Research (CIHR) [MOP-11-51-31; RFN 152985, 159815, 162303]
- Canadian Consortium of Neurodegeneration and Aging (CCNA; MOP-11-51-31 -team 1)
- Alzheimer’s Association [NIRG-12-92090, NIRP-12-259245], Brain Canada Foundation (CFI Project 34874; 33397)
- Fonds de Recherche du Québec – Santé (FRQS; Chercheur Boursier, 2020-VICO-279314)
- Swedish Alzheimer Foundation (grant #AF-940262)
- Alzheimer’s Association Research Fellowship (grant #AARFD-22-974564)
- University of Gothenburg
Collapse
Affiliation(s)
- Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - 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
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Ilaria Pola
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Joseph Therriault
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Stijn Servaes
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Jenna Stevenson
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jesse Klostranec
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Paolo Vitali
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Maxime Montembeault
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Cecile Tissot
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Arthur C Macedo
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - 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
| | - 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, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Pedro Rosa-Neto
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
16
|
Thal DR, Poesen K, Vandenberghe R, De Meyer S. Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers. Mol Neurodegener 2025; 20:33. [PMID: 40087672 PMCID: PMC11907863 DOI: 10.1186/s13024-025-00819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.
Collapse
Affiliation(s)
- Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.
| | - Koen Poesen
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Steffi De Meyer
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Antonioni A, Raho EM, Di Lorenzo F, Manzoli L, Flacco ME, Koch G. Blood phosphorylated Tau217 distinguishes amyloid-positive from amyloid-negative subjects in the Alzheimer's disease continuum. A systematic review and meta-analysis. J Neurol 2025; 272:252. [PMID: 40047958 PMCID: PMC11885345 DOI: 10.1007/s00415-025-12996-3] [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: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia worldwide, and cost-effective tools to detect amyloid pathology are urgently needed. Blood-based Tau phosphorylated at threonine 217 (pTau217) seems promising, but its reliability as a proxy for cerebrospinal fluid (CSF) status and ability to identify patients within the AD spectrum remain unclear. METHODS We performed a systematic review and meta-analysis on the potential of blood pTau217 to differentiate amyloid-positive (A+) and amyloid-negative (A-) subjects. We included original studies reporting quantitative data on pTau217 concentrations in both blood and CSF in the AD continuum. The single-group meta-analysis computed the pooled pTau217 levels in blood and in CSF, separately in the A+ and A- groups, while the head-to-head meta-analysis compared the mean pTau217 concentrations in the A+ versus A- subjects, both in blood and CSF, stratifying by assessment method in both cases. RESULTS Ten studies (819 A+; 1055 A-) were included. The mean pTau217 levels resulted higher in CSF than in blood and, crucially, in A+ individuals than in A- ones, regardless of the laboratory method employed. Most importantly, all laboratory techniques reliably distinguished A+ from A- subjects, whether applied to CSF or blood samples. CONCLUSIONS These results confirm that blood-based pTau217 is a reliable marker of amyloid pathology with significant implications for clinical practice in the AD continuum. Indeed, pTau217 might be a non-invasive, scalable biomarker for early AD detection, reducing the reliance on more invasive, expansive, and less accessible methods. CLINICAL TRIAL REGISTRATION Prospero CRD42024565187.
Collapse
Affiliation(s)
- Annibale Antonioni
- Doctoral Program in Translational Neurosciences and Neurotechnologies, Department of Neuroscience and Rehabilitation, University of Ferrara, Via Ludovico Ariosto, 35, 44121, Ferrara, Italy.
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy.
- Department of Neuroscience, Ferrara University Hospital, 44124, Ferrara, Italy.
| | - Emanuela Maria Raho
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
- Department of Neuroscience, Ferrara University Hospital, 44124, Ferrara, Italy
| | - Francesco Di Lorenzo
- Neuropsychophysiology Lab, Santa Lucia Foundation IRCCS, Via Ardeatina, 306, 00179, Rome, Italy.
| | - Lamberto Manzoli
- Department of Medical and Surgical Sciences, University of Bologna, 40126, Bologna, Italy
| | - Maria Elena Flacco
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Giacomo Koch
- Neuropsychophysiology Lab, Santa Lucia Foundation IRCCS, Via Ardeatina, 306, 00179, Rome, Italy.
- Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy.
- Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, 44121, Ferrara, Italy.
| |
Collapse
|
18
|
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: 1] [Impact Index Per Article: 1.0] [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
|
19
|
Cody KA, Du L, Studer RL, Jonaitis EM, Asthana S, Christian BT, Chin NA, Kirmess KM, Meyer MR, Yarasheski KE, West T, Verghese PB, Braunstein JB, Betthauser TJ, Langhough RE, Johnson SC. Accuracy of plasma biomarkers to detect Alzheimer's disease proteinopathy prior to dementia. Alzheimers Dement 2025; 21:e14570. [PMID: 40108829 PMCID: PMC11922805 DOI: 10.1002/alz.14570] [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: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Plasma biomarkers sensitive to Alzheimer's disease (AD) proteinopathy prior to the onset of dementia have significant implications for early detection. METHODS In 304 individuals without dementia, we investigated whether C2N Diagnostics' mass spectrometry (MS)-based plasma biomarkers (amyloid beta 42/40, %phosphorylated tau [p-tau]181, and %p-tau217) and amyloid probability scores (APS, PrecivityAD and APS2, PrecivityAD2) are associated with brain amyloid, brain tau, or preclinical cognitive decline. RESULTS In this cohort study, %p-tau217 and the APS2 had high discriminative accuracy (area under the curve > 0.93) for identifying elevated brain amyloid and tau and were associated with faster preclinical cognitive decline. Using %p-tau217 or the APS2 in a theoretical AD trial screening scenario reduced amyloid positron emission tomography imaging costs up to 41% or 45%, respectively. DISCUSSION These findings suggest that C2N Diagnostics' MS-based plasma biomarkers can detect brain amyloid and tau with high accuracy prior to dementia and could aid in identifying candidates for clinical trials or therapeutic intervention. HIGHLIGHTS C2N plasma biomarkers differentiated Alzheimer's disease proteinopathy status prior to dementia. Plasma %phosphorylated tau (p-tau)217 and the C2N Diagnostics PrecivityAD2 (APS2) were concordant with amyloid and tau positron emission tomography status. Plasma %p-tau217 and the APS2 were associated with preclinical cognitive decline.
Collapse
Affiliation(s)
- Karly A. Cody
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCaliforniaUSA
| | - Lianlian Du
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Rachel L. Studer
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Erin M. Jonaitis
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bradley T. Christian
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Waisman Laboratory for Brain Imaging and BehaviorUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Nathaniel A. Chin
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | | | | | | | - Tim West
- C2N DiagnosticsSt. LouisMissouriUSA
| | | | | | - Tobey J. Betthauser
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Rebecca E. Langhough
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| |
Collapse
|
20
|
Yun J, Shin D, Lee EH, Kim JP, Ham H, Gu Y, Chun MY, Kang SH, Kim HJ, Na DL, Kim CH, Kim KW, Kim SE, Kim Y, Kim J, Jung NY, Kim YJ, Cho SH, Zetterberg H, Blennow K, Gonzalez-Ortiz F, Ashton NJ, Therriault J, Rahmouni N, Rosa-Neto P, Weiner MW, Seo SW, Jang H. Temporal Dynamics and Biological Variability of Alzheimer Biomarkers. JAMA Neurol 2025:2830247. [PMID: 39960728 PMCID: PMC11833660 DOI: 10.1001/jamaneurol.2024.5263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/10/2024] [Indexed: 02/20/2025]
Abstract
Importance Understanding the characteristics of discordance between plasma biomarkers and positron emission tomography (PET) results in Alzheimer disease (AD) is crucial for accurate interpretation of the findings. Objective To compare (1) medical comorbidities affecting plasma biomarker concentrations, (2) imaging and clinical features, and (3) cognitive changes between plasma biomarker and PET discordant and concordant cases. Design, Setting, and Participants This multicenter cohort study, conducted between 2016 and 2023, included individuals with unimpaired cognition, mild cognitive impairment, or Alzheimer-type dementia, who had both amyloid β (Aβ) PET imaging and plasma biomarkers. A subset of participants also underwent tau PET imaging. Exposures Participants were categorized into 4 groups based on their plasma and PET biomarker results: plasma-/PET-, plasma+/PET-, plasma-/PET+, and plasma+/PET+. Main Outcomes and Measures Clinical characteristics were compared between the 4 groups, focusing on the discordant groups. Results A total of 2611 participants (mean [SD] age was 71.2 [8.7] years; 1656 female [63.4%]), of whom 124 additionally underwent tau PET, were included. Among the plasma biomarkers, phosphorylated tau (p-tau) 217 exhibited the highest concordance rate with Aβ (2326 of 2571 [90.5%]) and tau (100 of 120 [83.3%]) PET. The p-tau217+/Aβ PET- group was older (mean [SD] age, 75.8 [7.2] years vs 70.0 [8.8] years; P < .001) with a higher prevalence of hypertension (56 of 152 [36.8%] vs 266 of 1073 [25.0%]), diabetes (40 of 152 [26.3%] vs 156 of 1059 [14.7%]), and chronic kidney disease (17 of 152 [11.2%] vs 21 of 1073 [2.0%]) compared with the p-tau217-/Aβ PET- group (P < .001 for all). Body mass index was higher in p-tau217-/Aβ PET+ than in p-tau217+/Aβ PET+ (mean [SD], 24.1 [2.8] vs 23.1 [3.1], respectively; P = .001; calculated as weight in kilograms divided by height in meters squared). The p-tau217+/Aβ PET- group had lower hippocampal volume (mean [SD], 2555.4 [576.9] vs 2979.1 [545.8]; P < .001) and worse clinical trajectory compared with p-tau217-/Aβ PET- (β = -0.53; P < .001). In contrast, tau PET discordant cases did not show significant differences in medical comorbidities or clinical outcomes compared with the p-tau217-/tau PET- group. Only the p-tau 217+/tau PET+ group demonstrated faster cognitive deterioration compared with the p-tau 217-/tau PET- group (β = -1.66; P < .001). Conclusions and Relevance Results of this cohort study suggest that the mechanisms underlying the discordance between plasma biomarkers and PET findings may be multifaceted, underscoring the need to consider the temporal dynamics and biological variability of plasma biomarkers.
Collapse
Affiliation(s)
- Jihwan Yun
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
| | - Daeun Shin
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Eun Hye Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hongki Ham
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University. Seoul, South Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, South Korea
| | - Yuna Gu
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Min Young Chun
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University. Seoul, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Happymid Clinic, Seoul, South Korea
| | - Chi-Hun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Anyang, South Korea
| | - Ko Woon Kim
- Department of Neurology, Jeonbuk National University Medical School and Hospital, Jeonju, Korea
| | - Si Eun Kim
- Department of Neurology, Inje University College of Medicine, Haeundae Paik Hospital, Busan, Korea
| | - Yeshin Kim
- Department of Neurology, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Jaeho Kim
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Yeo Jin Kim
- Department of Neurology, Kangwon National University College of Medicine, Chuncheon-si, Korea
| | - Soo Hyun Cho
- Department of Neurology, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju, Korea
| | - 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, Gothenburg, 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, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792
| | - Kaj Blennow
- 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, Gothenburg, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People’s Republic of China
| | - Fernando Gonzalez-Ortiz
- 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, Gothenburg, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, United Kingdom
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Joseph Therriault
- McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Michael W. Weiner
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University. Seoul, South Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Hyemin Jang
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
VandeVrede L, Cho H, Sanderson-Cimino M, Wekselman F, Cobigo Y, Gorno-Tempini ML, Heuer HW, Kramer JH, Lario Lago A, Leichter D, Ljubenkov P, Miller BL, Perry DC, Rabinovici GD, Rojas JC, Rosen HJ, Saloner R, Staffaroni A, Triana-Baltzer G, Spina S, Seeley WW, Grinberg LT, Kolb HC, La Joie R, Boxer AL. Detection of Alzheimer Neuropathology in Alzheimer and Non-Alzheimer Clinical Syndromes With Blood-Based Biomarkers. JAMA Neurol 2025:2830013. [PMID: 39928343 PMCID: PMC11811866 DOI: 10.1001/jamaneurol.2024.5017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/22/2024] [Indexed: 02/11/2025]
Abstract
Importance Blood-based biomarkers for Alzheimer disease (AD) are clinically available, but their value is not well understood in syndromes typically associated with frontotemporal lobar degeneration syndromes (FTLD). Objective To investigate the clinical importance and detectability of AD in FTLD-related neurodegenerative syndromes using 3 plasma biomarkers, phosphorylated tau 217 (p-tau217), neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP). Design, Setting, and Participants This clinicopathological study took place at the University of California San Francisco Alzheimer Disease Research Center from August 2008 to July 2022. Autopsied individuals with clinical evaluation and neuropathological examination, diagnosed with clinical syndromes related to AD (n = 125), frontotemporal lobar degeneration (FTLD; n = 198), or cognitively unimpaired (CU) at the time of evaluation (n = 16) were included. Exposures AD-related or FTLD-related clinical syndromes or CU. Main Outcomes and Measures P-tau217, NfL, and GFAP were measured with single-molecule array (SIMOA). AD was defined as intermediate or high AD neuropathological change (ADNC) at autopsy. Clinical biomarker associations were evaluated using linear regressions. Imaging analyses used bayesian linear mixed-effects modeling. Results A total of 349 individuals (191 [55%] male; mean [SD] age at death, 72 [11] years) were included. AD was common in both AD-related syndromes (110/125 [88%]) and FTLD-related syndromes (45/198 [23%]). Neuropathological stage at autopsy was higher in AD-related syndromes (high ADNC: 82/88 [93%] AD vs 13/23 [56%] FTLD), and AD was frequently considered a copathology in FTLD-related syndromes (30/198 [15%]). Plasma p-tau217 concentrations were higher in AD-related syndromes (mean [SD], 0.28 [0.16] pg/mL) than FTLD-related syndromes (mean [SD], 0.10 [0.09] pg/mL) (P < .05). Plasma p-tau217 concentrations were highest in atypical AD-related syndromes (mean [SD], 0.33 [0.02] pg/mL), followed by typical late-onset amnestic syndromes (mean [SD], 0.27 [0.03] pg/mL). FTLD-related syndromes with AD (mean [SD], 0.19 [0.02] pg/mL) were higher compared to without (mean [SD], 0.07 [0.00] pg/mL). Plasma p-tau217 detected AD neuropathology across syndromes (area under the receiver operating characteristic curve [AUC], 0.95; 95% CI, 0.93-0.97), with slightly better performance in AD-related syndromes (AUC, 0.98; 95% CI, 0.95-1.00) compared to FTLD-related syndromes (AUC, 0.89; 95% CI, 0.83-0.94). NfL and GFAP had lower performance for detecting AD (AUC, 0.73; 95% CI, 0.68-0.78 and AUC, 0.75; 95% CI, 0.67-0.80, respectively) and added little to no diagnostic value either alone or in combinations with p-tau217. The presence of AD in FTLD-related syndromes was associated with lower Mini-Mental State Examination score (mean [SD], -2.90 [1.09]; P < .05), worse performance on memory (mean [SD] z score, -0.64 [0.32]), executive (mean [SD] z score, -0.74 [0.19]), and visuospatial composites (mean [SD] z score, -0.88 [0.37]) as well as increased rates of posterior cortical atrophy. Conclusion Clinically relevant AD was prevalent across neurodegenerative syndromes and detectable with plasma p-tau217. Plasma p-tau217 may be a useful tool to investigate the clinical impact of AD copathology in non-AD neurodegenerative syndromes, including the effect of disease-modifying therapies.
Collapse
Affiliation(s)
- Lawren VandeVrede
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Hanna Cho
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mark Sanderson-Cimino
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Fattin Wekselman
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Yann Cobigo
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Hilary W. Heuer
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Joel H. Kramer
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Argentina Lario Lago
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Dana Leichter
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Peter Ljubenkov
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Bruce L. Miller
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - David C. Perry
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Gil D. Rabinovici
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
- Associate Editor, JAMA Neurology
| | - Julio C. Rojas
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Howard J. Rosen
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Rowan Saloner
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Adam Staffaroni
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | | | - Salvatore Spina
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - William W. Seeley
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Lea T. Grinberg
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
- Department of Pathology, University of California, San Francisco
| | - Hartmuth C. Kolb
- Neuroscience Biomarkers, Janssen Research & Development, San Diego, California
| | - Renaud La Joie
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Adam L. Boxer
- Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| |
Collapse
|
22
|
Mielke MM, Evans JK, Neiberg RH, Molina-Henry DP, Marcovina SM, Johnson KC, Carmichael OT, Rapp SR, Sachs BC, Ding J, Shappell HM, Luchsinger JA, Espeland MA, Hayden KM. Alzheimer Disease Blood Biomarkers and Cognition Among Individuals With Diabetes and Overweight or Obesity. JAMA Netw Open 2025; 8:e2458149. [PMID: 39913137 PMCID: PMC11803481 DOI: 10.1001/jamanetworkopen.2024.58149] [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: 09/16/2024] [Accepted: 11/27/2024] [Indexed: 02/07/2025] Open
Abstract
Importance Blood-based biomarkers (BBMs) are clinically available to aid in the diagnosis of Alzheimer disease (AD) and AD-related dementias (ADRD), but their association with cognition among older adults with specific chronic conditions has not been examined. Objective To longitudinally examine associations between baseline AD and ADRD BBMs and change in BBMs with cognition among participants with type 2 diabetes (T2D) and overweight or obesity. Design, Setting, and Participants The Look AHEAD (Action for Health in Diabetes) study was a clinical trial of older adults with T2D and overweight or obesity randomized to a 10-year intensive lifestyle intervention for weight loss or a diabetes support and education condition. Participants were recruited and followed up at 16 clinical sites across the US. Enrollment occurred from January 1, 2001, to December 31, 2004. The primary intervention spanned the first 4 years after participants' enrollment (January 1, 2008, to December 31, 2011). The clinical trial was stopped in September 2012 and was converted to an observational study. Blood samples were drawn at baseline and 8 to 12 years later. Cognitive assessments were performed from January 1, 2013, to December 31, 2014, and from January 1, 2018, to December 31, 2020. Data for the present cohort study were analyzed between January and August 2024. Exposures Baseline and 8- to 12-year change in plasma levels of amyloid-β (Aβ)40, Aβ42, Aβ42/40 ratio, phosphorylated tau 181 (pTau-181), glial fibrillary acidic protein (GFAP), and neurofilament light chain (NfL). Main Outcomes and Measures Cognitive composite z score and adjudicated mild cognitive impairment or probable dementia. Results The mean (SD) baseline age of 758 participants was 61.5 (6.1) years, and 424 participants [55.9%] were female. Mean (SD) body mass index was 34.8 (5.3). Of the participants, 373 were randomized to diabetes support and education and 385 to intensive lifestyle intervention. Increasing baseline BBM levels were not associated with any cognitive composite z score. Increasing levels of NfL (β = -0.032 [SE, 0.013]; P = .01) and GFAP (β = -0.087 [SE, 0.025]; P < .001), but not the Aβ42/40 ratio (β = 0.006 [SE, 0.040]; P = .88) or pTau-181 (β = 0.026 [SE, 0.025]; P = .31), were associated with worsening cognitive function and incident mild cognitive impairment or probable dementia. The intervention had no association with 8- to 12-year change in BBM levels. Conclusions and Relevance In this study of participants with T2D and overweight or obesity, increasing plasma NfL and GFAP levels over time, but not Aβ42/40 or pTau-181 levels, were associated with cognitive decline and incident cognitive impairment. These results suggest that plasma NfL and GFAP may be important biomarkers of cognitive change among this patient population.
Collapse
Affiliation(s)
- Michelle M. Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Joni K. Evans
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Rebecca H. Neiberg
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Doris P. Molina-Henry
- Winston-Salem State University, Winston-Salem, North Carolina
- Alzheimer’s Therapeutic Research Institute, Keck School of Medicine, University of Southern California, Los Angeles
| | | | - Karen C. Johnson
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis
| | - Owen T. Carmichael
- Biomedical Imaging Center, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Stephen R. Rapp
- Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, North Carolina
- Department of Psychiatry and Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Bonnie C. Sachs
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Division of Gerontology and Geriatric Medicine Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Jingzhong Ding
- Department of Internal Medicine, Division of Gerontology and Geriatric Medicine Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Heather M. Shappell
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Jose A. Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, New York
| | - Mark A. Espeland
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Division of Gerontology and Geriatric Medicine Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Kathleen M. Hayden
- Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
23
|
Ashton NJ, Keshavan A, Brum WS, Andreasson U, Arslan B, Droescher M, Barghorn S, Vanbrabant J, Lambrechts C, Van Loo M, Stoops E, Iyengar S, Ji H, Xu X, Forrest‐Hay A, Zhang B, Luo Y, Jeromin A, Vandijck M, Bastard NL, Kolb H, Triana‐Baltzer G, Bali D, Janelidze S, Yang S, Demos C, Romero D, Sigal G, Wohlstadter J, Malyavantham K, Khare M, Jethwa A, Stoeckl L, Gobom J, Kac PR, Gonzalez‐Ortiz F, Montoliu‐Gaya L, Hansson O, Rissman RA, Carrillo MC, Shaw LM, Blennow K, Schott JM, Zetterberg H. The Alzheimer's Association Global Biomarker Standardization Consortium (GBSC) plasma phospho-tau Round Robin study. Alzheimers Dement 2025; 21:e14508. [PMID: 39907496 PMCID: PMC11851162 DOI: 10.1002/alz.14508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 02/06/2025]
Abstract
INTRODUCTION The Alzheimer's Association Global Biomarker Standardization Consortium conducted a blinded case-control study to learn which phosphorylated tau (p-tau) assays provide the largest fold-changes in Alzheimer's disease (AD) versus non-AD and show commutability in measuring patient samples and candidate certified reference materials (CRMs). METHODS Thirty-three different p-tau assays measured paired plasma and cerebrospinal fluid (CSF) from 40 participants (25 with "AD pathology" and 15 with "non-AD pathology" by CSF amyloid beta [Aβ]42/Aβ40 and p-tau181 criteria). Four CRMs were assessed. RESULTS Plasma p-tau217 demonstrated higher fold-changes between AD and non-AD than other p-tau epitopes. Fujirebio LUMIPULSE G, UGOT IPMS, and Lilly MSD p-tau217 provided the highest fold-changes. Plasma p-tau217 showed the strongest correlations between plasma assays (rho = 0.81-0.97). The CRMs were not commutable across assays. DISCUSSION Plasma p-tau217 showed larger fold-changes and better accuracy for detecting AD pathology in symptomatic individuals, with greater cross-platform agreement than other p-tau variants. Further work is needed to develop suitable CRMs facilitating cross-assay standardization. HIGHLIGHTS Paired plasma and cerebrospinal fluid (CSF) samples from twenty-five Alzheimer's disease (AD) and 15 non-AD patients were measured blind. Thirty-three plasma assays were compared, for phosphorylated tau-181 (p-tau181), 205, 212, 217 and 231. Plasma p-tau217 consistently had the highest fold-change and was best correlated between assays. Plasma-CSF correlations were weak to moderate. There was lack of commutability for four candidate reference materials.
Collapse
|
24
|
Jang H, Shin D, Yoo H, Zetterberg H, Blennow K, Gonzalez‐Ortiz F, Ashton NJ, Day TA, Lee EH, Yun J, Na DL, Kim HJ, Kang SH, Kim KW, Kim SE, Kim YJ, Kim Y, Kim J, Kim C, Chun MY, Jung NY, Cho SH, Kim JP, Seo SW, the K‐ROAD study groups. Differential roles of Alzheimer's disease plasma biomarkers in stepwise biomarker-guided diagnostics. Alzheimers Dement 2025; 21:e14526. [PMID: 39907189 PMCID: PMC11848384 DOI: 10.1002/alz.14526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 02/06/2025]
Abstract
INTRODUCTION This study aimed to investigate the differential roles of various plasma biomarkers in a stepwise diagnostic strategy for Alzheimer's disease (AD). METHODS A total of 2984 participants, including 666 cognitively unimpaired (CU), 2032 with Alzheimer's clinical syndrome (ACS), and 286 non-ACS individuals, were recruited. Plasma amyloid beta (Aβ) 42/40, four phosphorylated tau (p-tau) epitopes, glial fibrillary acidic protein (GFAP), and neurofilament light chain (NfL) levels were measured using immunoassays. RESULTS NfL demonstrated fair to excellent accuracy in differentiating non-ACS from CU groups (area under the curve [AUC], 0.79 to 0.94). p-tau217 had the highest accuracy for identifying Aβ (AUC 0.94) and tau positron emission tomography status (AUC 0.91). In the ACS group, p-tau217 was the strongest predictor of cognitive decline (p < .001). DISCUSSION NfL may serve as a useful screening tool, while p-tau217 is particularly valuable for confirming AD pathology and prognosis. HIGHLIGHTS Plasma NfL could screen for cognitive impairment. p-tau217 reliably detects AD pathology, regardless of diagnosis. p-tau217 and GFAP predict prognosis in ACS. Each plasma biomarker plays a distinct role in stepwise AD diagnostics.
Collapse
Grants
- RS-2020-KH106434 Korea Dementia Research Center
- RS-2020-KH107436 Korea Dementia Research Center
- 2024-ER1003-00 Korea National Institute of Health
- #SMX1240561 Future Medicine 20*30 Project of the Samsung Medical Center
- RS-2019-NR040057 National Research Foundation of Korea (NRF)
- RS-2021-II212068 Institute of Information & communications Technology Planning & Evaluation (IITP)
- RS-2022-KH125667 Korea Health Industry Development Institute (KHIDI)
- 2024-R001 Korean Dementia Association
- 2023-00356 Swedish Research Council
- 2022-01018 Swedish Research Council
- 2022-00732 Swedish Research Council
- 2019-02397 Swedish Research Council
- 2017-00915 Swedish Research Council
- 101053962 European Union Horizon Europe research and innovation program
- ALFGBG-71320 Swedish State Support for Clinical Research
- ALFGBG-715986 Swedish State Support for Clinical Research
- ALFGBG-965240 Swedish State Support for Clinical Research
- 201809-2016862 Alzheimer Drug Discovery Foundation (ADDF)
- ADSF-21-831376-C AD Strategic Fund and the Alzheimer's Association
- ADSF-21-831381-C AD Strategic Fund and the Alzheimer's Association
- ADSF-21-831377-C AD Strategic Fund and the Alzheimer's Association
- ADSF-24-1284328-C AD Strategic Fund and the Alzheimer's Association
- FO2022-0270 Bluefield Project, Cure Alzheimer's Fund, Olav Thon Foundation, Erling-Persson Family Foundation, Familjen Rönströms Stiftelse, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- FO2017-0243 Bluefield Project, Cure Alzheimer's Fund, Olav Thon Foundation, Erling-Persson Family Foundation, Familjen Rönströms Stiftelse, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- ALZ2022-0006 Bluefield Project, Cure Alzheimer's Fund, Olav Thon Foundation, Erling-Persson Family Foundation, Familjen Rönströms Stiftelse, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- 860197 European Union's Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement
- JPND2021-00694 European Union Joint Programme - Neurodegenerative Disease Research
- JPND2019-466-236 European Union Joint Programme - Neurodegenerative Disease Research
- UKDRI-1003 National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, and the UK Dementia Research Institute at UCL
- AF-930351 Swedish Alzheimer Foundation
- AF-939721 Swedish Alzheimer Foundation
- AF-968270 Swedish Alzheimer Foundation
- AF-994551 Swedish Alzheimer Foundation
- ZEN-21-848495 Alzheimer's Association 2021 Zenith Award
- SG-23-1038904QC Alzheimer's Association 2022-2025 Grant
- La Fondation Recherche Alzheimer (FRA), Paris, France, Kirsten and Freddy Johansen Foundation, Copenhagen, Denmark, and Familjen Rönströms Stiftelse, Stockholm, Sweden.
- Korea Dementia Research Center
- Korea National Institute of Health
- Swedish Research Council
Collapse
|
25
|
Datta D, Arnsten AFT. The etiology and prevention of early-stage tau pathology in higher cortical circuits: Insights from aging rhesus macaques. Alzheimers Dement 2025; 21:e14477. [PMID: 39776253 PMCID: PMC11848412 DOI: 10.1002/alz.14477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Aging rhesus macaques provide a unique model for learning how age and inflammation drive early-stage pathology in sporadic Alzheimer's disease, and for testing potential therapeutics. Unlike mice, aging macaques have extensive association cortices and inflammatory signaling similar to humans, are apolipoprotein E ε4 homozygotes, and naturally develop tau and amyloid pathology with marked cognitive deficits. Importantly, monkeys provide the unique opportunity to study early-stage, soluble hyperphosphorylated tau (p-tau), including p-tau217. As soluble p-tau is rapidly dephosphorylated post mortem, it is not captured in human brains except with biopsy material. However, new macaque data show that soluble p-tau is toxic to neurons and capable of seeding across cortical circuits. Extensive evidence indicates that age-related inflammatory signaling contributes to calcium dysregulation, which drives tau hyperphosphorylation and amyloid beta generation. Pharmacological studies in aged macaques suggest that inhibiting inflammation and restoring calcium regulation can reduce tau hyperphosphorylation with minimal side effects, appropriate for potential preventive therapeutics. HIGHLIGHTS: Aging monkeys provide a unique window into early stage, soluble phosphorylated tau (p-tau). Inflammation with advancing age leads to calcium dysregulation, p-tau, and amyloid beta (Aβ). Macaque research shows p-tau undergoes transsynaptic seeding early in the cortex. p-tau traps amyloid precursor protein-containing endosomes, which may increase Aβ and drive vicious cycles. Restoring calcium regulation in cortex reduced p-tau217 levels in aged macaques.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of PsychiatryYale Medical SchoolNew HavenConnecticutUSA
| | - Amy F. T. Arnsten
- Department of NeuroscienceYale Medical SchoolNew HavenConnecticutUSA
| |
Collapse
|
26
|
Sarto J, Esteller-Gauxax D, Guillén N, Falgàs N, Borrego-Écija S, Massons M, Fernández-Villullas G, González Y, Tort-Merino A, Bosch B, Castellví M, Piñol-Ripoll G, Juncà-Parella J, Del Val A, Pérez-Millan A, Comas A, Antonell A, Naranjo L, Ruiz-García R, Augé JM, Sánchez-Valle R, Lladó A, Balasa M. Accuracy and clinical applicability of plasma tau 181 and 217 for Alzheimer's disease diagnosis in a memory clinic cohort. J Neurol 2025; 272:160. [PMID: 39849125 DOI: 10.1007/s00415-025-12897-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025]
Abstract
Plasma tau phosphorylated at threonine 181 (p-tau181) and 217 (p-tau217) have demonstrated high accuracy for Alzheimer's disease (AD) diagnosis, defined by CSF/PET amyloid beta (Aβ) positivity, but most studies have been performed in research cohorts, limiting their generalizability. We studied plasma p-tau217 and p-tau181 for CSF Aβ status discrimination in a cohort of consecutive patients attending an academic memory clinic in Spain (July 2019-June 2024). All patients had CSF AD biomarkers performed as part of their routine clinical assessment. Aβ positivity was defined with a local cut-off of CSF Aβ1-42 < 600 pg/mL; in patients with borderline Aβ1-42 values or when there was a mismatch between the Aβ and the T status (T + if CSF p-tau181 ≥ 65 pg/mL), a ratio Aβ1-42/Aβ1-40 < 0.07 was used. Plasma p-tau217 and p-tau181 were measured retrospectively, from blood samples collected at first visit, with Fujirebio Lumipulse and Quanterix Simoa assays, respectively. We included 468 patients (mean age 67 years, 50% female, 61% Aβ positive). Plasma p-tau217 outperformed plasma p-tau181 in discriminating CSF Aβ status (AUC 0.95 vs 0.90, p = 0.005). A 97.5% sensitivity and specificity plasma p-tau217 algorithm, classifying patients into three groups of Aβ probability (Low, Intermediate and High), resulted in 67% of patients in the Low and High groups, having their Aβ status predicted (as negative and positive, respectively) with 96% accuracy. The remaining 33% in the Intermediate group were candidates to undergo CSF/PET testing. A model with a 10% variation in p-tau217 levels yielded small changes in accuracy (95%). In conclusion, plasma p-tau217 could have discriminated CSF Aβ status in two-thirds of patients with very high accuracy in a memory clinic cohort. These results support the implementation of plasma p-tau217 as an initial diagnostic tool in memory clinics for AD diagnosis, reducing the need for more invasive/expensive testing.
Collapse
Affiliation(s)
- Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Diana Esteller-Gauxax
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
| | - Núria Guillén
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Miquel Massons
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
| | - Guadalupe Fernández-Villullas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Yolanda González
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
| | - Adrià Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Magda Castellví
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
| | - Gerard Piñol-Ripoll
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Unitat Trastorns Cognitius, Cognition and Behaviour Study Group, Santa Maria University Hospital, IRBLleida, Lleida, Spain
| | - Jordi Juncà-Parella
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Andrea Del Val
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
| | - Agnès Pérez-Millan
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Aina Comas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laura Naranjo
- Immunology Service, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raquel Ruiz-García
- Immunology Service, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Josep María Augé
- Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Villaroel 170, 08036, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
27
|
Kjaergaard D, Simonsen AH, Waldemar G, Nielsen TR. Ethnic and racial influences on blood biomarkers for Alzheimer's disease: A systematic review. J Alzheimers Dis 2025; 103:81-91. [PMID: 39814528 DOI: 10.1177/13872877241299047] [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: 01/18/2025]
Abstract
BACKGROUND Little is known about confounding factors influencing Alzheimer's disease (AD) blood biomarker concentrations. OBJECTIVE The objective of this systematic review was to explore the available evidence for the influences of ethnicity and race on AD blood biomarker concentrations. METHODS We conducted a comprehensive systematic search in PubMed and Web of Science databases spanning from inception until 15 June 2023. We included studies that utilized plasma or serum biomarkers (amyloid-β [Aβ], total tau [t-tau], phosphorylated tau [p-tau], neurofilament light [NfL], and glial fibrillary acidic protein [GFAP]), compared individuals with AD to healthy controls, and included a minimum of two ethnic or racial groups for comparison. A total of 10 studies were included in the qualitative synthesis. All studies were conducted in the US. RESULTS Seven studies reported differences in blood biomarker concentrations between ethnic or racial groups. However, after adjusting for medical conditions and social determinants of health, the differences became non-significant in two of the studies. The included studies differed in their included covariates and their statistical approaches, which complicated the interpretation of the observed differences. CONCLUSIONS The available evidence suggests that ethnicity and race may influence blood biomarker concentrations. However, it remains unclear to what extent these differences are mediated by differences in social determinants of health and medical conditions. Future studies are needed to explore ethnic and racial differences in blood biomarkers, including studies in diverse samples outside the US.
Collapse
Affiliation(s)
- Daniel Kjaergaard
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anja Hviid Simonsen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - T Rune Nielsen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Lee EH, Kang SH, Shin D, Kim YJ, Zetterberg H, Blennow K, Gonzalez‐Ortiz F, Ashton NJ, Cheon BK, Yoo H, Ham H, Yun J, Kim JP, Kim HJ, Na DL, Jang H, Seo SW, the K‐ROAD study group. Plasma Alzheimer's disease biomarker variability: Amyloid-independent and amyloid-dependent factors. Alzheimers Dement 2025; 21:e14368. [PMID: 39535473 PMCID: PMC11782842 DOI: 10.1002/alz.14368] [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: 07/09/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION We aimed to investigate which factors affect plasma biomarker levels via amyloid beta (Aβ)-independent or Aβ-dependent effects and improve the predictive performance of these biomarkers for Aβ positivity on positron emission tomography (PET). METHODS A total of 2935 participants underwent blood sampling for measurements of plasma Aβ42/40 ratio, phosphorylated tau 217 (p-tau217; ALZpath), glial fibrillary acidic protein (GFAP), and neurofilament light chain (NfL) levels using single-molecule array and Aβ PET. Laboratory findings were collected using a routine blood test battery. RESULTS Aβ-independent factors included hemoglobin and estimated glomerular filtration rate (eGFR) for p-tau217 and hemoglobin, eGFR, and triiodothyronine (T3) for GFAP and NfL. Aβ-dependent factors included apolipoprotein E genotypes, body mass index status for Aβ42/40, p-tau217, GFAP, and NfL. However, these factors exhibited negligible or modest effects on Aβ positivity on PET. DISCUSSION Our findings highlight the importance of accurately interpreting plasma biomarkers for predicting Aβ uptake in real-world settings. HIGHLIGHTS We investigated factor-Alzheimer's disease plasma biomarker associations in a large Korean cohort. Hemoglobin and estimated glomerular filtration rate affect the biomarkers independently of brain amyloid beta (Aβ). Apolipoprotein E genotypes and body mass index status affect the biomarkers dependent on brain Aβ. Addition of Aβ-independent factors shows negligible effect in predicting Aβ positivity. Adjusting for Aβ-dependent factors shows a modest effect in predicting Aβ positivity.
Collapse
|
29
|
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
|
30
|
Martínez-Dubarbie F, Guerra-Ruiz A, López-García S, Lage C, Fernández-Matarrubia M, Pozueta-Cantudo A, García-Martínez M, Corrales-Pardo A, Bravo M, López-Hoyos M, Irure-Ventura J, de Lucas EM, Drake-Pérez M, García-Unzueta MT, Sánchez-Juan P, Rodríguez-Rodríguez E. Longitudinal trajectory of plasma p-tau217 in cognitively unimpaired subjects. Alzheimers Res Ther 2024; 16:268. [PMID: 39702464 PMCID: PMC11661039 DOI: 10.1186/s13195-024-01642-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The advent of Alzheimer's disease-modifying drugs requires accurate biological diagnosis to identify candidates for these therapies. So far, the most promising single plasma biomarker is phosphorylated tau at threonine 217 (p-tau217). To understand its biological features, it is essential to know its longitudinal trajectory and factors influencing it in cognitively unimpaired subjects with no brain pathology. METHODS We analyzed longitudinal plasma p-tau217 values (mean follow-up time = 768.3 days) in a cohort of 209 healthy volunteers. We have studied factors associated with plasma p-tau217 changes by using different linear mixed-effects models. RESULTS In amyloid-negative cognitively healthy subjects (n = 151) carriers of ApoE ε4 allele had significantly higher p-tau217 values than non-carriers (0.85 pg/mL; p-value < 0.001) and also a greater rate of change (0.01 pg/mL/year; p-value < 0.001). In the overall sample, including subjects with amyloid and tau pathology we have seen that amyloid positive subjects had higher predicted baseline plasma p-tau217 values than amyloid negative subjects (0.16 pg/mL; p-value < 0.001) and a greater rate of change (0.00004 pg/mL/day; p-value < 0.001). Subjects considered tau positive also showed a greater rate of change of p-tau217 with respect to tau negative (0.00005 pg/mL/day; p-value < 0.001). A + T + N + participants showed a higher baseline p-tau217 levels than A-T-N- subjects (0.2 pg/mL; p-value < 0.001) and also a greater rate of change (0.00006 pg/mL/day; p-value = 0.002). ApoE ε4 carriers had a greater rate of change than non-carriers (0.00003 pg/mL/day; p-value = 0.03). CONCLUSION In amyloid-negative cognitively unimpaired subjects, ApoE4 status influenced both baseline levels and rate of change of plasma p-tau217. Other factors such as age, sex or glomerular filtration rate have not shown significant influence on plasma p-tau217 levels in this group.
Collapse
Affiliation(s)
- Francisco Martínez-Dubarbie
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain.
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain.
- Network Center for Biomedical Research in Neurodegenerative Diseases, CIBERNED, National Institute of Health Carlos III, Madrid, 28220, Spain.
- , Avda. de Valdecilla 25, Santander, Cantabria, 39008, Spain.
| | - Armando Guerra-Ruiz
- Biochemistry and Clinical Analysis Department, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
| | - Sara López-García
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - Carmen Lage
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases, CIBERNED, National Institute of Health Carlos III, Madrid, 28220, Spain
- Atlantic Fellow for Equity in Brain health, Global Brain Health Institute, University of California, San Francisco, 94158, USA
| | - Marta Fernández-Matarrubia
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases, CIBERNED, National Institute of Health Carlos III, Madrid, 28220, Spain
| | - Ana Pozueta-Cantudo
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - María García-Martínez
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - Andrea Corrales-Pardo
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - María Bravo
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
| | - Marcos López-Hoyos
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Immunology Department, Marqués de Valdecilla University Hospital, Santander, 39008, Spain
- Molecular Biology Department, University of Cantabria, Santander, 39011, Spain
| | - Juan Irure-Ventura
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Immunology Department, Marqués de Valdecilla University Hospital, Santander, 39008, Spain
| | - Enrique Marco de Lucas
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Radiology Department, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Marta Drake-Pérez
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Radiology Department, Marqués de Valdecilla University Hospital, Santander, Spain
| | - María Teresa García-Unzueta
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Biochemistry and Clinical Analysis Department, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
| | - Pascual Sánchez-Juan
- Network Center for Biomedical Research in Neurodegenerative Diseases, CIBERNED, National Institute of Health Carlos III, Madrid, 28220, Spain
- Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, Madrid, 28031, Spain
| | - Eloy Rodríguez-Rodríguez
- Neurology Service, Marqués de Valdecilla University Hospital, Santander, Cantabria, 39008, Spain
- Institute for Research Marqués de Valdecilla (IDIVAL), Santander, Cantabria, 39011, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases, CIBERNED, National Institute of Health Carlos III, Madrid, 28220, Spain
- Medicine and Psychiatry Department, University of Cantabria, Santander, 39011, Spain
| |
Collapse
|
31
|
Bathla S, Datta D, Bolat D, Woo E, Duque A, Arellano J, Arnsten A, Nairn AC. Dysregulated calcium signaling in the aged macaque entorhinal cortex associated with tau hyperphosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.626721. [PMID: 39713378 PMCID: PMC11661118 DOI: 10.1101/2024.12.05.626721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Tau pathology in sporadic Alzheimer's disease (AD) follows a distinct pattern, beginning in the entorhinal cortex (ERC) and spreading to interconnected brain regions. Early-stage tau pathology, characterized by soluble phosphorylated tau, is difficult to study in human brains post-mortem due to rapid dephosphorylation. Rhesus macaques, which naturally develop age-related tau pathology resembling human AD, provide an ideal model for investigating early tau etiology. This study examines the molecular processes underlying tau pathology in the macaque ERC, focusing on calcium and inflammatory signaling pathways. Our findings reveal age-related decreases in PDE4 phosphodiesterases that hydrolyze cAMP and increases in calpain-2 and GCPII that occur in parallel with early-stage tau hyperphosphorylation at multiple epitopes (pS214-tau, pT181-tau, pT217-tau). These findings suggest that dysregulated calcium signaling in ERC, beginning in middle-age, primes tau for hyperphosphorylation, potentially driving the early stages of AD, advancing our understanding of how ERC vulnerabilities contribute to neurodegeneration in AD.
Collapse
|
32
|
Göschel L, Dell'Orco A, Fillmer A, Aydin S, Ittermann B, Riemann L, Lehmann S, Cano S, Melin J, Pendrill L, Hoede PL, Teunissen CE, Schwarz C, Grittner U, Körtvélyessy P, Flöel A. Plasma p-tau181 and GFAP reflect 7T MR-derived changes in Alzheimer's disease: A longitudinal study of structural and functional MRI and MRS. Alzheimers Dement 2024; 20:8684-8699. [PMID: 39558898 DOI: 10.1002/alz.14318] [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: 04/25/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Associations between longitudinal changes of plasma biomarkers and cerebral magnetic resonance (MR)-derived measurements in Alzheimer's disease (AD) remain unclear. METHODS In a study population (n = 127) of healthy older adults and patients within the AD continuum, we examined associations between longitudinal plasma amyloid beta 42/40 ratio, tau phosphorylated at threonine 181 (p-tau181), glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL), and 7T structural and functional MR imaging and spectroscopy using linear mixed models. RESULTS Increases in both p-tau181 and GFAP showed the strongest associations to 7T MR-derived measurements, particularly with decreasing parietal cortical thickness, decreasing connectivity of the salience network, and increasing neuroinflammation as determined by MR spectroscopy (MRS) myo-inositol. DISCUSSION Both plasma p-tau181 and GFAP appear to reflect disease progression, as indicated by 7T MR-derived brain changes which are not limited to areas known to be affected by tau pathology and neuroinflammation measured by MRS myo-inositol, respectively. HIGHLIGHTS This study leverages high-resolution 7T magnetic resonance (MR) imaging and MR spectroscopy (MRS) for Alzheimer's disease (AD) plasma biomarker insights. Tau phosphorylated at threonine 181 (p-tau181) and glial fibrillary acidic protein (GFAP) showed the largest changes over time, particularly in the AD group. p-tau181 and GFAP are robust in reflecting 7T MR-based changes in AD. The strongest associations were for frontal/parietal MR changes and MRS neuroinflammation.
Collapse
Affiliation(s)
- Laura Göschel
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andrea Dell'Orco
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Berlin, Germany
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ariane Fillmer
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Berlin, Germany
| | - Semiha Aydin
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Berlin, Germany
| | - Layla Riemann
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Berlin, Germany
- Institute for Applied Medical Informatics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
| | | | - Jeanette Melin
- Division Safety and Transport, Division Measurement Science and Technology, RISE, Research Institutes of Sweden, Gothenburg, Sweden
| | - Leslie Pendrill
- Division Safety and Transport, Division Measurement Science and Technology, RISE, Research Institutes of Sweden, Gothenburg, Sweden
| | - Patty L Hoede
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Claudia Schwarz
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Ulrike Grittner
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Péter Körtvélyessy
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Standort Magdeburg, Magdeburg, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Standort Rostock/Greifswald, Greifswald, Germany
| |
Collapse
|
33
|
Phillips JM, Winfree RL, Seto M, Schneider JA, Bennett DA, Dumitrescu LC, Hohman TJ. Pathologic and clinical correlates of region-specific brain GFAP in Alzheimer's disease. Acta Neuropathol 2024; 148:69. [PMID: 39580758 PMCID: PMC11586308 DOI: 10.1007/s00401-024-02828-5] [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: 07/26/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
Plasma glial fibrillary acidic protein (GFAP) is an emerging biomarker of Alzheimer's disease (AD), with higher blood GFAP levels linked to faster cognitive decline, particularly among individuals with high brain amyloid burden. However, few studies have examined brain GFAP expression to clarify if peripheral associations reflect brain changes. This study aimed to correlate region-specific GFAP mRNA expression (n = 917) and protein abundance (n=386) with diverse neuropathological measures at autopsy in the Religious Orders Study and Rush Memory and Aging Project (ROS/MAP) and to characterize the interaction between brain GFAP and brain amyloid burden on downstream outcomes. We assessed GFAP gene expression in the dorsolateral prefrontal cortex, caudate nucleus, and posterior cingulate cortex with respect to core AD pathology (amyloid-β and tau), cerebrovascular (microinfarcts, macroinfarcts, and cerebral amyloid angiopathy [CAA]), proteinopathic (TDP-43, Lewy bodies), and cognitive outcomes. These associations were further examined at the protein level using tandem-mass tag proteomic measurements from the dorsolateral prefrontal cortex. We also assessed GFAP interactions with AD neuropathology on downstream outcomes. Cortical GFAP gene and protein expression were significantly upregulated in participants with a neuropathologically confirmed AD diagnosis at autopsy (all PFDR < 3.5e-4), but not in individuals positive for tau pathology and negative for amyloid pathology (all PFDR > 0.05). Higher cortical GFAP levels were associated with increased amyloid pathology, CAA pathology, and faster cognitive decline (all PFDR < 3.3e-3). GFAP's associations with phosphorylated tau burden and cognition were influenced by amyloid burden, being most pronounced among amyloid-positive individuals, confirming previous in vivo biomarker observations. No associations were observed between GFAP gene expression and outcomes in the caudate nucleus. Our results support previous biomarker findings and suggest that higher brain GFAP levels are associated with higher brain amyloid burden and faster cognitive decline among amyloid-positive individuals.
Collapse
Affiliation(s)
- Jared M Phillips
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rebecca L Winfree
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mabel Seto
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Logan C Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
34
|
Kovacech B, Cullen NC, Novak P, Hanes J, Kontsekova E, Katina S, Parrak V, Fresser M, Vanbrabant J, Feldman HH, Winblad B, Stoops E, Vanmechelen E, Zilka N. Post hoc analysis of ADAMANT, a phase 2 clinical trial of active tau immunotherapy with AADvac1 in patients with Alzheimer's disease, positive for plasma p-tau217. Alzheimers Res Ther 2024; 16:254. [PMID: 39580468 PMCID: PMC11585249 DOI: 10.1186/s13195-024-01620-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The spread of tau pathology closely correlates with the disease course and cognitive decline in Alzheimer's disease (AD). Tau-targeting immunotherapies are being developed to stop the spread of tau pathology and thus halt disease progression. In this post hoc analysis of the ADAMANT clinical trial, we examined the performance of AADvac1, an active immunotherapy targeting the microtubule-binding region (MTBR) of tau, in a subgroup of participants with elevated plasma p-tau217, indicating AD-related neuropathological changes. METHODS ADAMANT was a 24-month, randomized, placebo-controlled, parallel-group, double-blinded, multicenter, phase 2 clinical trial in subjects with mild AD. The trial participants were randomized 3:2 to receive six doses of AADvac1 or placebo at 4-week intervals, followed by five booster doses at 14-week intervals. The primary outcome was safety. The secondary outcomes were the Clinical Dementia Rating-Sum of Boxes (CDR-SB), the Alzheimer's Disease Cooperative Study - Activities of Daily Living score for Mild Cognitive Impairment 18-item version (ADCS-ADL-MCI-18), and immunogenicity. Volumetric MRI, plasma neurofilament light (NfL), and glial fibrillary acidic protein (GFAP) were exploratory outcomes. The inclusion criterion for this post-hoc analysis was a baseline plasma p-tau217 level above the cutoff for AD. RESULTS Among 196 ADAMANT participants, 137 were positive for plasma p-tau217 (mean age 71.4 years, 59% women). AADvac1 was safe and well tolerated in this subgroup. AADvac1 reduced the rate of accumulation of log-plasma NfL by 56% and that of GFAP by 73%. The treatment differences in the CDR-SB and ADCS-ADL-MCI-18 scores favored AADvac1 but were not statistically significant. AADvac1 had no effect on whole-brain volume but nonsignificantly reduced the loss of brain cortical tissue in several regions. Importantly, the impact on the study outcomes was more pronounced in participants with higher anti-tau antibody levels. CONCLUSIONS These results suggest that AADvac1 tau immunotherapy can reduce plasma biomarkers of neurodegeneration and neuroinflammation. These findings and possible observations on brain atrophy and cognition are hypothesis-generating and warrant further evaluation in a larger clinical trial. TRIAL REGISTRATION EudraCT 2015-000630-30 (primary) and NCT02579252.
Collapse
Affiliation(s)
- Branislav Kovacech
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia.
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Clinical Research Centre, Jan Waldenströms Gata 35, 202 13, Malmö, Sweden
| | - Petr Novak
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Jozef Hanes
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Eva Kontsekova
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102 Bratislava, Slovakia and Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava, 84510, Slovakia
| | - Stanislav Katina
- Department of Mathematics and Statistics, Axon Neuroscience R&D Services SE, Bratislava, Slovakia, and (current) Masaryk University, Kotlářská 267/2, Brno, 611 37, Czech Republic
| | - Vojtech Parrak
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia
| | - Michal Fresser
- Axon Neuroscience SE, 4 Arch. Makariou & Kalogreon, 6016, Larnaca, Cyprus
| | | | - Howard H Feldman
- Department of Neurosciences, Alzheimer's Disease Cooperative Study, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, 171 64, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 86, Stockholm, Sweden
| | - Erik Stoops
- ADx NeuroSciences NV, Technologiepark 6, 9052, Ghent, Belgium
| | | | - Norbert Zilka
- Axon Neuroscience R&D Services SE, Dvorakovo Nabr. 10, 81102, Bratislava, Slovakia.
| |
Collapse
|
35
|
Hofmann A, Häsler LM, Lambert M, Kaeser SA, Gräber-Sultan S, Obermüller U, Kuder-Buletta E, la Fougere C, Laske C, Vöglein J, Levin J, Fox NC, Ryan NS, Zetterberg H, Llibre-Guerra JJ, Perrin RJ, Ibanez L, Schofield PR, Brooks WS, Day GS, Farlow MR, Allegri RF, Chrem Mendez P, Ikeuchi T, Kasuga K, Lee JH, Roh JH, Mori H, Lopera F, Bateman RJ, McDade E, Gordon BA, Chhatwal JP, Jucker M, Schultz SA. Comparative neurofilament light chain trajectories in CSF and plasma in autosomal dominant Alzheimer's disease. Nat Commun 2024; 15:9982. [PMID: 39557867 PMCID: PMC11574007 DOI: 10.1038/s41467-024-52937-8] [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: 04/11/2024] [Accepted: 09/25/2024] [Indexed: 11/20/2024] Open
Abstract
Disease-modifying therapies for Alzheimer's disease (AD) are likely to be most beneficial when initiated in the presymptomatic phase. To track the benefit of such interventions, fluid biomarkers are of great importance, with neurofilament light chain protein (NfL) showing promise for monitoring neurodegeneration and predicting cognitive outcomes. Here, we update and complement previous findings from the Dominantly Inherited Alzheimer Network Observational Study by using matched cross-sectional and longitudinal cerebrospinal fluid (CSF) and plasma samples from 567 individuals, allowing timely comparative analyses of CSF and blood trajectories across the entire disease spectrum. CSF and plasma trajectories were similar at presymptomatic stages, discriminating mutation carriers from non-carrier controls 10-20 years before the estimated onset of clinical symptoms, depending on the statistical model used. However, after symptom onset the rate of change in CSF NfL continued to increase steadily, whereas the rate of change in plasma NfL leveled off. Both plasma and CSF NfL changes were associated with grey-matter atrophy, but not with Aβ-PET changes, supporting a temporal decoupling of Aβ deposition and neurodegeneration. These observations support NfL in both CSF and blood as an early marker of neurodegeneration but suggest that NfL measured in the CSF may be better suited for monitoring clinical trial outcomes in symptomatic AD patients.
Collapse
Affiliation(s)
- Anna Hofmann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lisa M Häsler
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Marius Lambert
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stephan A Kaeser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | - Ulrike Obermüller
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | - Christian la Fougere
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig Maximilians-Universität München, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig Maximilians-Universität München, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Henrik Zetterberg
- Department Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jorge J Llibre-Guerra
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura Ibanez
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter R Schofield
- Neuroscience Research Australia, Randwick, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - William S Brooks
- Neuroscience Research Australia, Randwick, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health Sydney, University of New South Wales, Sydney, Australia
| | - Gregory S Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Martin R Farlow
- Indiana Alzheimer Disease Center and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Takeshi Ikeuchi
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Kensaku Kasuga
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jee Hoon Roh
- Departments of Neurology and Physiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Hiroshi Mori
- Faculty of Medicine, Osaka Metropolitan University, Nagaoka Sutoku University, Osaka, Japan
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia (GNA), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
- Brigham and Women's Hospital Boston, Boston, MA, USA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| | - Stephanie A Schultz
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
36
|
Miner AE, Groh JR, Tripodis Y, Adler CH, Balcer LJ, Bernick C, Zetterberg H, Blennow K, Peskind E, Ashton NJ, Gaudet CE, Martin B, Palmisano JN, Banks SJ, Barr WB, Wethe JV, Cantu RC, Dodick DW, Katz DI, Mez J, van Amerongen S, Cummings JL, Shenton ME, Reiman EM, Stern RA, Alosco ML, for the DIAGNOSE CTE Research Project. Examination of plasma biomarkers of amyloid, tau, neurodegeneration, and neuroinflammation in former elite American football players. Alzheimers Dement 2024; 20:7529-7546. [PMID: 39351900 PMCID: PMC11567811 DOI: 10.1002/alz.14231] [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: 03/14/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Blood-based biomarkers offer a promising approach for the detection of neuropathologies from repetitive head impacts (RHI). We evaluated plasma biomarkers of amyloid, tau, neurodegeneration, and inflammation in former football players. METHODS The sample included 180 former football players and 60 asymptomatic, unexposed male participants (aged 45-74). Plasma assays were conducted for beta-amyloid (Aβ) 40, Aβ42, hyper-phosphorylated tau (p-tau) 181+231, total tau (t-tau), neurofilament light (NfL), glial fibrillary acidic protein (GFAP), interleukin-6 (IL-6), Aβ42/p-tau181 and Aβ42/Aβ40 ratios. We evaluated their ability to differentiate the groups and associations with RHI proxies and traumatic encephalopathy syndrome (TES). RESULTS P-tau181 and p-tau231(padj = 0.016) were higher and Aβ42/p-tau181 was lower(padj = 0.004) in football players compared to controls. Discrimination accuracy for p-tau was modest (area under the curve [AUC] = 0.742). Effects were not attributable to AD-related pathology. Younger age of first exposure (AFE) correlated with higher NfL (padj = 0.03) and GFAP (padj = 0.033). Plasma GFAP was higher in TES-chronic traumatic encephalopathy (TES-CTE) Possible/Probable (padj = 0.008). DISCUSSION Plasma p-tau181 and p-tau231, GFAP, and NfL may offer some usefulness for the characterization of RHI-related neuropathologies. HIGHLIGHTS Former football players had higher plasma p-tau181 and p-tau231 and lower Aβ42/ptau-181 compared to asymptomatic, unexposed men. Younger age of first exposure was associated with increased plasma NfL and GFAP in older but not younger participants. Plasma GFAP was higher in participants with TES-CTE possible/probable compared to TES-CTE no/suggestive.
Collapse
Grants
- ZEN-21-848495 Alzheimer's Association 2021 Zenith Award
- ALZ2022-0006 Hjärnfonden, Sweden
- U01 NS093334 NINDS NIH HHS
- ALFGBG-965240 Hjärnfonden, Sweden
- JPND2021-00694 European Union Joint Programme-Neurodegenerative Disease Research
- UKDRI-1003 UK Dementia Research Institute at UCL
- 2022-00732 UK Dementia Research Institute at UCL
- SG-23-1038904 QC Alzheimer's Association 2022-2025 Grant
- AF-939721 Swedish Alzheimer Foundation
- AF-930351 Swedish Alzheimer Foundation
- RF1 NS132290 NINDS NIH HHS
- AF-994551 Swedish Alzheimer Foundation
- ADSF-21-831381-C AD Strategic Fund and the Alzheimer's Association
- Bluefield Project, Cure Alzheimer's Fund
- JPND2019-466-236 European Union Joint Program for Neurodegenerative Disorders
- 2017-00915 UK Dementia Research Institute at UCL
- Olav Thon Foundation, the Erling-Persson Family Foundation
- FO2017-0243 Hjärnfonden, Sweden
- ADSF-21-831376-C AD Strategic Fund and the Alzheimer's Association
- European Union's Horizon 2020
- ADSF-24-1284328-C AD Strategic Fund and the Alzheimer's Association
- RF1NS132290 National Institute of Neurological Disorders and Stroke/National Institute on Aging
- Kirsten and Freddy Johansen Foundation, Copenhagen, Denmark
- ALFGBG-715986 Hjärnfonden, Sweden
- #ALFGBG-71320 Swedish State Support for Clinical Research
- AF-968270 Swedish Alzheimer Foundation
- ADSF-21-831377-C AD Strategic Fund and the Alzheimer's Association
- FO2022-0270 Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- 101053962 European Union's Horizon Europe
- 201809-2016862 Alzheimer Drug Discovery Foundation
- La Fondation Recherche Alzheimer
- U01NS093334 National Institute of Neurological Disorders and Stroke (NINDS)
- National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre
Collapse
|
37
|
Therriault J, Janelidze S, Benedet AL, Ashton NJ, Arranz Martínez J, Gonzalez-Escalante A, Bellaver B, Alcolea D, Vrillon A, Karim H, Mielke MM, Hyung Hong C, Roh HW, Contador J, Puig Pijoan A, Algeciras-Schimnich A, Vemuri P, Graff-Radford J, Lowe VJ, Karikari TK, Jonaitis E, Brum W, Tissot C, Servaes S, Rahmouni N, Macedo AC, Stevenson J, Fernandez-Arias J, Wang YT, Woo MS, Friese MA, Jia WL, Dumurgier J, Hourregue C, Cognat E, Ferreira PL, Vitali P, Johnson S, Pascoal TA, Gauthier S, Lleó A, Paquet C, Petersen RC, Salmon D, Mattsson-Carlgren N, Palmqvist S, Stomrud E, Galasko D, Son SJ, Zetterberg H, Fortea J, Suárez-Calvet M, Jack CR, Blennow K, Hansson O, Rosa-Neto P. Diagnosis of Alzheimer's disease using plasma biomarkers adjusted to clinical probability. NATURE AGING 2024; 4:1529-1537. [PMID: 39533113 PMCID: PMC11564087 DOI: 10.1038/s43587-024-00731-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 09/25/2024] [Indexed: 11/16/2024]
Abstract
Recently approved anti-amyloid immunotherapies for Alzheimer's disease (AD) require evidence of amyloid-β pathology from positron emission tomography (PET) or cerebrospinal fluid (CSF) before initiating treatment. Blood-based biomarkers promise to reduce the need for PET or CSF testing; however, their interpretation at the individual level and the circumstances requiring confirmatory testing are poorly understood. Individual-level interpretation of diagnostic test results requires knowledge of disease prevalence in relation to clinical presentation (clinical pretest probability). Here, in a study of 6,896 individuals evaluated from 11 cohort studies from six countries, we determined the positive and negative predictive value of five plasma biomarkers for amyloid-β pathology in cognitively impaired individuals in relation to clinical pretest probability. We observed that p-tau217 could rule in amyloid-β pathology in individuals with probable AD dementia (positive predictive value above 95%). In mild cognitive impairment, p-tau217 interpretation depended on patient age. Negative p-tau217 results could rule out amyloid-β pathology in individuals with non-AD dementia syndromes (negative predictive value between 90% and 99%). Our findings provide a framework for the individual-level interpretation of plasma biomarkers, suggesting that p-tau217 combined with clinical phenotyping can identify patients where amyloid-β pathology can be ruled in or out without the need for PET or CSF confirmatory testing.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Javier Arranz Martínez
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Armand Gonzalez-Escalante
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Agathe Vrillon
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Helmet Karim
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Michelle M Mielke
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Chang Hyung Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyun Woong Roh
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - José Contador
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
| | - Albert Puig Pijoan
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- ERA-Net on Cardiovascular Diseases Consortium, Barcelona, Spain
| | | | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Erin Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Wagner Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidad Federal do RioGrande do Sul, Porto Alegre, Brazil
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Marcel S Woo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wan Lu Jia
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Julien Dumurgier
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Claire Hourregue
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Emmanuel Cognat
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | | | - Paolo Vitali
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sterling Johnson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Claire Paquet
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Ronald C Petersen
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - David Salmon
- San Diego and Shiley-Marcos Alzheimer's Disease Research Center, University of California, La Jolla, CA, USA
| | - 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
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmo, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmo, Sweden
| | - Douglas Galasko
- San Diego and Shiley-Marcos Alzheimer's Disease Research Center, University of California, La Jolla, CA, USA
| | - Sang Joon Son
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- ERA-Net on Cardiovascular Diseases Consortium, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain
| | - Marc Suárez-Calvet
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
38
|
Pichet Binette A, Gaiteri C, Wennström M, Kumar A, Hristovska I, Spotorno N, Salvadó G, Strandberg O, Mathys H, Tsai LH, Palmqvist S, Mattsson-Carlgren N, Janelidze S, Stomrud E, Vogel JW, Hansson O. Proteomic changes in Alzheimer's disease associated with progressive Aβ plaque and tau tangle pathologies. Nat Neurosci 2024; 27:1880-1891. [PMID: 39187705 PMCID: PMC11452344 DOI: 10.1038/s41593-024-01737-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 07/23/2024] [Indexed: 08/28/2024]
Abstract
Proteomics can shed light on the dynamic and multifaceted alterations in neurodegenerative disorders like Alzheimer's disease (AD). Combining radioligands measuring β-amyloid (Aβ) plaques and tau tangles with cerebrospinal fluid proteomics, we uncover molecular events mirroring different stages of AD pathology in living humans. We found 127 differentially abundant proteins (DAPs) across the AD spectrum. The strongest Aβ-related proteins were mainly expressed in glial cells and included SMOC1 and ITGAM. A dozen proteins linked to ATP metabolism and preferentially expressed in neurons were independently associated with tau tangle load and tau accumulation. Only 20% of the DAPs were also altered in other neurodegenerative diseases, underscoring AD's distinct proteome. Two co-expression modules related, respectively, to protein metabolism and microglial immune response encompassed most DAPs, with opposing, staggered trajectories along the AD continuum. We unveil protein signatures associated with Aβ and tau proteinopathy in vivo, offering insights into complex neural responses and potential biomarkers and therapeutics targeting different disease stages.
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| | - Chris Gaiteri
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
- Rush University Alzheimer's Disease Center, Rush University, Chicago, IL, USA
| | - Malin Wennström
- Cognitive Disorder Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Atul Kumar
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Ines Hristovska
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Hansruedi Mathys
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- University of Pittsburgh Brain Institute and Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Rush University Alzheimer's Disease Center, Rush University, Chicago, IL, USA
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Jacob W Vogel
- Department of Clinical Sciences Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
39
|
Jack CR, Graf A, Burnham SC, Doty EG, Moebius HJ, Montenigro P, Siemers E, Sink KM, Shaw LM, Hansen CT, Wildsmith KR, Mahinrad S, Carrillo MC, Weber CJ. Application of the revised criteria for diagnosis and staging of Alzheimer's disease: Drug development and clinical practice. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e70013. [PMID: 39748835 PMCID: PMC11694534 DOI: 10.1002/trc2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 01/04/2025]
Abstract
The newly proposed revised criteria for diagnosis and staging of Alzheimer's disease (AD) by the Alzheimer's Association (AA) Workgroup represent a significant milestone in the field. These criteria offer objective measures for diagnosing and staging biological AD, bridging the gap between research and clinical care. Although implementation feasibility may vary across regions and settings, improving the availability and accuracy of biomarkers, especially plasma biomarkers, is expected to enhance the applicability of these criteria in clinical practice. The Fall 2023 Alzheimer's Association Research Roundtable (AARR) meeting served as a forum for gathering industry perspectives and feedback on these revised criteria, ensuring that the new criteria inform research, clinical trial design, and clinical care. In this article, we outline a summary of the newly proposed "Revised Criteria for Diagnosis and Staging of AD: AA Workgroup" and provide highlights from the AARR meeting in fall 2023. Highlights The Alzheimer's Association Research Roundtable (AARR) convened leaders from industry, academia, and government, to review the Revised Criteria for Diagnosis and Staging of AD: AA Workgroup, and gather industry perspectives and feedback on these revised criteria before its publication.The newly proposed revised criteria for diagnosis and staging of Alzheimer's disease (AD) by the AA's Workgroup represent a significant milestone, offering objective measures for the biological and staging of AD and bridging the gap between research and clinical care.Improving the availability and accuracy of biomarkers, especially blood-based biomarkers (BBMs) is expected to improve clinical research and enhance the applicability of these criteria in clinical practice.
Collapse
Affiliation(s)
| | - Ana Graf
- Novartis Pharma AGBaselSwitzerland
| | | | | | | | | | | | | | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of Medicine University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | | | | | | | | |
Collapse
|
40
|
Abdelnour C, Young CB, Shahid-Besanti M, Smith A, Wilson EN, Benitez JR, Vossler H, Plastini MJ, Winer JR, Kerchner GA, Cholerton B, Andreasson KI, Henderson VW, Yutsis M, Montine TJ, Tian L, Mormino EC, Poston KL. Plasma pTau181 Reveals a Pathological Signature that Predicts Cognitive Outcomes in Lewy Body Disease. Ann Neurol 2024; 96:526-538. [PMID: 38888142 PMCID: PMC11324388 DOI: 10.1002/ana.27003] [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: 12/03/2023] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVE To determine whether plasma phosphorylated-Tau181 (pTau181) could be used as a diagnostic biomarker of concurrent Alzheimer's disease neuropathologic change (ADNC) or amyloidosis alone, as well as a prognostic, monitoring, and susceptibility/risk biomarker for clinical outcomes in Lewy body disease (LBD). METHODS We studied 565 participants: 94 LBD with normal cognition, 83 LBD with abnormal cognition, 114 with Alzheimer's disease, and 274 cognitively normal. Plasma pTau181 levels were measured with the Lumipulse G platform. Diagnostic accuracy for concurrent ADNC and amyloidosis was assessed with Receiver Operating Characteristic curves in a subset of participants with CSF pTau181/Aβ42, and CSF Aβ42/Aβ40 or amyloid-β PET, respectively. Linear mixed effects models were used to examine the associations between baseline and longitudinal plasma pTau181 levels and clinical outcomes. RESULTS Plasma pTau181 predicted concurrent ADNC and amyloidosis in LBD with abnormal cognition with 87% and 72% accuracy, respectively. In LBD patients with abnormal cognition, higher baseline plasma pTau181 was associated with worse baseline MoCA and CDR-SB, as well as accelerated decline in CDR-SB. Additionally, in this group, rapid increases in plasma pTau181 over 3 years predicted a faster decline in CDR-SB and memory. In LBD patients with normal cognition, there was no association between baseline or longitudinal plasma pTau181 levels and clinical outcomes; however, elevated pTau181 at baseline increased the risk of conversion to cognitive impairment. INTERPRETATION Our findings suggest that plasma pTau181 is a promising biomarker for concurrent ADNC and amyloidosis in LBD. Furthermore, plasma pTau181 holds potential as a prognostic, monitoring, and susceptibility/risk biomarker, predicting disease progression in LBD. ANN NEUROL 2024;96:526-538.
Collapse
Affiliation(s)
- Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Marian Shahid-Besanti
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alena Smith
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Edward N. Wilson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Javier Ramos Benitez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hillary Vossler
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Melanie J. Plastini
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph R. Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey A. Kerchner
- Pharma Research and Early Development, F. Hoffmann-La Roche, Ltd., Roche, Basel, Switzerland
| | - Brenna Cholerton
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Victor W. Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Maya Yutsis
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lu Tian
- Department of Biomedical Data Science, School of Medicine. Department of Statistics, School of Humanities and Sciences. Stanford University, CA, USA
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen L. Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
41
|
Ashton NJ, Keshavan A, Brum WS, Andreasson U, Arslan B, Droescher M, Barghorn S, Vanbrabant J, Lambrechts C, Van Loo M, Stoops E, Iyengar S, Ji H, Xu X, Forrest-Hay A, Zhang B, Luo Y, Jeromin A, Vandijck M, Bastard NL, Kolb H, Triana-Baltzer G, Bali D, Janelidze S, Yang SY, Demos C, Romero D, Sigal G, Wohlstadter J, Malyavantham K, Khare M, Jethwa A, Stoeckl L, Gobom J, Kac PR, Gonzalez-Ortiz F, Montoliu-Gaya L, Hansson O, Rissman RA, Carillo MC, Shaw LM, Blennow K, Schott JM, Zetterberg H. The Alzheimer's Association Global Biomarker Standardization Consortium (GBSC) plasma phospho-tau Round Robin study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.22.24312244. [PMID: 39228740 PMCID: PMC11370527 DOI: 10.1101/2024.08.22.24312244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
BACKGROUND Phosphorylated tau (p-tau) is a specific blood biomarker for Alzheimer's disease (AD) pathology. Multiple p-tau biomarkers on several analytical platforms are poised for clinical use. The Alzheimer's Association Global Biomarker Standardisation Consortium plasma phospho-tau Round Robin study engaged assay developers in a blinded case-control study on plasma p-tau, aiming to learn which assays provide the largest fold-changes in AD compared to non-AD, have the strongest relationship between plasma and cerebrospinal fluid (CSF), and show the most consistent relationships between methods (commutability) in measuring both patient samples and candidate reference materials (CRM). METHODS Thirty-three different p-tau biomarker assays, built on eight different analytical platforms, were used to quantify paired plasma and CSF samples from 40 participants. AD biomarker status was categorised as "AD pathology" (n=25) and "non-AD pathology" (n=15) by CSF Aβ42/Aβ40 (US-FDA; CE-IVDR) and p-tau181 (CE-IVDR) methods. The commutability of four CRM, at three concentrations, was assessed across assays. FINDINGS Plasma p-tau217 consistently demonstrated higher fold-changes between AD and non-AD pathology groups, compared to other p-tau epitopes. Fujirebio LUMIPULSE G, UGOT IPMS, and Lilly MSD p-tau217 assays provided the highest median fold-changes. In CSF, p-tau217 assays also performed best, and exhibited substantially larger fold-changes than their plasma counterparts, despite similar diagnostic performance. P-tau217 showed the strongest correlations between plasma assays (rho=0.81 to 0.97). Plasma p-tau levels were weakly-to-moderately correlated with CSF p-tau, and correlations were non-significant within the AD group alone. The evaluated CRM were not commutable across assays. INTERPRETATION Plasma p-tau217 measures had larger fold-changes and discriminative accuracies for detecting AD pathology, and better agreement across platforms than other plasma p-tau variants. Plasma and CSF markers of p-tau, measured by immunoassays, are not substantially correlated, questioning the interchangeability of their continuous relationship. Further work is warranted to understand the pathophysiology underlying this dissociation, and to develop suitable reference materials facilitating cross-assay standardisation. FUNDING Alzheimer's Association (#ADSF-24-1284328-C).
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 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
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Ashvini Keshavan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Mathias Droescher
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Stefan Barghorn
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | | | | | - Maxime Van Loo
- ADx NeuroSciences N.V., Technologiepark 6, 9052 Ghent, Belgium
| | - Erik Stoops
- ADx NeuroSciences N.V., Technologiepark 6, 9052 Ghent, Belgium
| | | | - HaYeun Ji
- Alamar Biosciences, Inc., Fremont, CA, USA
| | - Xiaomei Xu
- Alamar Biosciences, Inc., Fremont, CA, USA
| | | | | | - Yuling Luo
- Alamar Biosciences, Inc., Fremont, CA, USA
| | | | | | | | | | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, California, USA
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund 22184, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund 22184, Sweden
| | | | | | - Daniel Romero
- Meso Scale Diagnostics, LLC., Rockville, Maryland, USA
| | - George Sigal
- Meso Scale Diagnostics, LLC., Rockville, Maryland, USA
| | | | | | | | | | | | - Johan Gobom
- 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
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund 22184, Sweden
- Memory Clinic, Skåne University Hospital, Malmö 20502, Sweden
| | - Robert A Rissman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA 92121, USA
| | - Maria C Carillo
- Division of Medical & Scientific Relations, Alzheimer's Association, Chicago, Illinois, USA
| | - Leslie M Shaw
- Department of pathology & laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Memory Clinic, Skåne University Hospital, Malmö 20502, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Memory Clinic, Skåne University Hospital, Malmö 20502, Sweden
- UK Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Science Park, Hong Kong, China
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
42
|
Bader I, Groot C, Tan HS, Milongo JMA, Haan JD, Verberk IMW, Yong K, Orellina J, Campbell S, Wilson D, van Harten AC, Kok PHB, van der Flier WM, Pijnenburg YAL, Barkhof F, van de Giessen E, Teunissen CE, Bouwman FH, Ossenkoppele R. Rationale and design of the BeyeOMARKER study: prospective evaluation of blood- and eye-based biomarkers for early detection of Alzheimer's disease pathology in the eye clinic. Alzheimers Res Ther 2024; 16:190. [PMID: 39169442 PMCID: PMC11340081 DOI: 10.1186/s13195-024-01545-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a common, complex and multifactorial disease that may require screening across multiple routes of referral to enable early detection and subsequent future implementation of tailored interventions. Blood- and eye-based biomarkers show promise as low-cost, scalable and patient-friendly tools for early AD detection given their ability to provide information on AD pathophysiological changes and manifestations in the retina, respectively. Eye clinics provide an intriguing real-world proof-of-concept setting to evaluate the performance of these potential AD screening tools given the intricate connections between the eye and brain, presumed enrichment for AD pathology in the aging population with eye disorders, and the potential for an accelerated diagnostic pathway for under-recognized patient groups. METHODS The BeyeOMARKER study is a prospective, observational, longitudinal cohort study aiming to include individuals visiting an eye-clinic. Inclusion criteria entail being ≥ 50 years old and having no prior dementia diagnosis. Excluded eye-conditions include traumatic insults, superficial inflammation, and conditions in surrounding structures of the eye that are not engaged in vision. The BeyeOMARKER cohort (n = 700) will undergo blood collection to assess plasma p-tau217 levels and a brief cognitive screening at the eye clinic. All participants will subsequently be invited for annual longitudinal follow-up including remotely administered cognitive screening and questionnaires. The BeyeOMARKER + cohort (n = 150), consisting of 100 plasma p-tau217 positive participants and 50 matched negative controls selected from the BeyeOMARKER cohort, will additionally undergo Aβ-PET and tau-PET, MRI, retinal imaging including hyperspectral imaging (primary), widefield imaging, optical coherence tomography (OCT) and OCT-Angiography (secondary), and cognitive and cortical vision assessments. RESULTS We aim to implement the current protocol between April 2024 until March 2027. Primary outcomes include the performance of plasma p-tau217 and hyperspectral retinal imaging to detect AD pathology (using Aβ- and tau-PET visual read as reference standard) and to detect cognitive decline. Initial follow-up is ~ 2 years but may be extended with additional funding. CONCLUSIONS We envision that the BeyeOMARKER study will demonstrate the feasibility of early AD detection based on blood- and eye-based biomarkers in alternative screening settings, and will improve our understanding of the eye-brain connection. TRIAL REGISTRATION The BeyeOMARKER study (Eudamed CIV ID: CIV-NL-23-09-044086; registration date: 19th of March 2024) is approved by the ethical review board of the Amsterdam UMC.
Collapse
Affiliation(s)
- Ilse Bader
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands.
| | - Colin Groot
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - H Stevie Tan
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC Location VUmc, Amsterdam Reproduction and Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Jean-Marie A Milongo
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Jurre den Haan
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Keir Yong
- Queen Square Institute of Neurology, Dementia Research Centre, London, WC1N 3BG, UK
| | | | | | | | - Argonde C van Harten
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Pauline H B Kok
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Wiesje M van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Yolande A L Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Frederik Barkhof
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
- UCL Queen Square Institute of Neurology and Centre for Medical Image Computing, University College, London, WC1N 3BG, UK
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Femke H Bouwman
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Rik Ossenkoppele
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
| |
Collapse
|
43
|
Lai R, Li B, Bishnoi R. P-tau217 as a Reliable Blood-Based Marker of Alzheimer's Disease. Biomedicines 2024; 12:1836. [PMID: 39200300 PMCID: PMC11351463 DOI: 10.3390/biomedicines12081836] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Amyloid plaques and tau tangles are the hallmark pathologic features of Alzheimer's disease (AD). Traditionally, these changes are identified in vivo via cerebrospinal fluid (CSF) analysis or positron emission tomography (PET) scans. However, these methods are invasive, expensive, and resource-intensive. To address these limitations, there has been ongoing research over the past decade to identify blood-based markers for AD. Despite the challenges posed by their extremely low concentrations, recent advances in mass spectrometry and immunoassay techniques have made it feasible to detect these blood markers of amyloid and tau deposition. Phosphorylated tau (p-tau) has shown greater promise in reflecting amyloid pathology as evidenced by CSF and PET positivity. Various isoforms of p-tau, distinguished by their differential phosphorylation sites, have been recognized for their ability to identify amyloid-positive individuals. Notable examples include p-tau181, p-tau217, and p-tau235. Among these, p-tau217 has emerged as a superior and reliable marker of amyloid positivity and, thus, AD in terms of accuracy of diagnosis and ability for early prognosis. In this narrative review, we aim to elucidate the utility of p-tau217 as an AD marker, exploring its underlying basis, clinical diagnostic potential, and relevance in clinical care and trials.
Collapse
Affiliation(s)
- Roy Lai
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA (B.L.)
| | - Brenden Li
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA (B.L.)
| | - Ram Bishnoi
- Department of Psychiatry and Behavioral Neurosciences, University of South Florida, Tampa, FL 33613, USA
- USF Health Byrd Alzheimer’s Center and Research Institute, Tampa, FL 33613, USA
- USF Memory Disorder Clinic, Tampa, FL 33613, USA
| |
Collapse
|
44
|
Garnier-Crussard A. Association between treatment with sacubitril/valsartan and the risk of Alzheimer's disease: a clinical update. Alzheimers Res Ther 2024; 16:177. [PMID: 39090680 PMCID: PMC11293045 DOI: 10.1186/s13195-024-01547-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
Since 2014, sacubitril/valsartan (Entresto®) is widely prescribed for heart failure. Despite neprilysin inhibition's benefits in heart failure, concerns about potential amyloid-beta (Aβ) accumulation and Alzheimer's disease (AD) risk have persisted. This narrative review, a decade post-approval, evaluates the risk of amyloid pathology and neurocognitive disorders in long-term sacubitril/valsartan use. Clinical trials, real-world studies, and pharmacovigilance data do not indicate an increased risk of cognitive decline. In patients treated with sacubitril/valsartan blood-based amyloid biomarkers show perturbations, while neuroimaging biomarkers reveal no significant increase in amyloid load. Despite a theoretical risk of amyloid accumulation and AD under treatment with sacubitril/valsartan, current clinical data appears reassuring, and there is no signal indicating an increased risk of cognitive decline, but a perturbation of amyloid blood-based biomarkers, which implies great caution when interpreting biomarkers in this context.
Collapse
Affiliation(s)
- Antoine Garnier-Crussard
- Clinical and Research Memory Centre of Lyon, Lyon Institute For Aging, Hospices Civils de Lyon, Hôpital des Charpennes, 27 rue Gabriel Péri, Villeurbanne, 69100, France.
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Neuropresage Team, Cyceron, Caen, 14000, France.
| |
Collapse
|
45
|
Nguyen Ho PT, Hoepel SJW, Rodriguez-Ayllon M, Luik AI, Vernooij MW, Neitzel J. Sleep, 24-Hour Activity Rhythms, and Subsequent Amyloid-β Pathology. JAMA Neurol 2024; 81:824-834. [PMID: 38913396 PMCID: PMC11197458 DOI: 10.1001/jamaneurol.2024.1755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/15/2024] [Indexed: 06/25/2024]
Abstract
Importance Sleep disturbances are common among older adults and have been associated with the development of Alzheimer disease (AD), such as amyloid-β (Aβ) pathology. For effective AD prevention, it is essential to pinpoint the specific disturbances in sleep and the underlying 24-hour activity rhythms that confer the highest risk of Aβ deposition. Objective To determine the associations of 24-hour activity rhythms and sleep with Aβ deposition in adults without dementia, to evaluate whether disrupted 24-hour activity and sleep may precede Aβ deposition, and to assess the role of the apolipoprotein E ε4 (APOE4) genotype. Design, Setting, and Participants This was an observational cohort study using data from the Rotterdam Study. Of 639 participants without dementia who underwent Aβ positron emission tomography (PET) from September 2018 to November 2021, 319 were included in the current study. Exclusion criteria were no APOE genotyping and no valid actigraphy data at the baseline visits from 2004 to 2006 or from 2012 to 2014. The mean (SD) follow-up was 7.8 (2.4) years. Data were analyzed from March 2023 to April 2024. Exposures Actigraphy (7 days and nights, objective sleep, and 24-hour activity rhythms), sleep diaries (self-reported sleep), Aβ42/40, phosphorylated tau (p-tau)181 and p-tau217 plasma assays, 18F-florbetaben PET (mean standard uptake value ratio [SUVR] in a large cortical region of interest), and APOE4 genotype. Main Outcomes and Measures Association of objective and self-reported sleep and 24-hour activity rhythms at baseline with brain Aβ PET burden at follow-up. Results The mean (range) age in the study population was 61.5 (48-80) years at baseline and 69.2 (60-88) years at follow-up; 150 (47%) were women. Higher intradaily variability at baseline, an indicator of fragmented 24-hour activity rhythms, was associated with higher Aβ PET burden at follow-up (β, 0.15; bootstrapped 95% CI, 0.04 to 0.26; bootstrapped P = .02, false discovery rate [FDR] P = .048). APOE genotype modified this association, which was stronger in APOE4 carriers (β, 0.38; bootstrapped 95% CI, 0.05 to 0.64; bootstrapped P = .03) compared to noncarriers (β, 0.07; bootstrapped 95% CI, -0.04 to 0.18; bootstrapped P = .19). The findings remained largely similar after excluding participants with AD pathology at baseline, suggesting that a fragmented 24-hour activity rhythm may have preceded Aβ deposition. No other objective or self-reported measure of sleep was associated with Aβ. Conclusions and Relevance Among community-dwelling adults included in this study, higher fragmentation of the 24-hour activity rhythms was associated with greater subsequent Aβ burden, especially in APOE4 carriers. These results suggest that rest-activity fragmentation could represent a modifiable risk factor for AD.
Collapse
Affiliation(s)
- Phuong Thuy Nguyen Ho
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Sanne J. W. Hoepel
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Maria Rodriguez-Ayllon
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Annemarie I. Luik
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Trimbos Institute—the Netherlands Institute of Mental Health and Addiction, Utrecht, the Netherlands
| | - Meike W. Vernooij
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Julia Neitzel
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
46
|
Jack CR, Andrews JS, Beach TG, Buracchio T, Dunn B, Graf A, Hansson O, Ho C, Jagust W, McDade E, Molinuevo JL, Okonkwo OC, Pani L, Rafii MS, Scheltens P, Siemers E, Snyder HM, Sperling R, Teunissen CE, Carrillo MC. Revised criteria for diagnosis and staging of Alzheimer's disease: Alzheimer's Association Workgroup. Alzheimers Dement 2024; 20:5143-5169. [PMID: 38934362 PMCID: PMC11350039 DOI: 10.1002/alz.13859] [Citation(s) in RCA: 509] [Impact Index Per Article: 509.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 06/28/2024]
Abstract
The National Institute on Aging and the Alzheimer's Association convened three separate work groups in 2011 and single work groups in 2012 and 2018 to create recommendations for the diagnosis and characterization of Alzheimer's disease (AD). The present document updates the 2018 research framework in response to several recent developments. Defining diseases biologically, rather than based on syndromic presentation, has long been standard in many areas of medicine (e.g., oncology), and is becoming a unifying concept common to all neurodegenerative diseases, not just AD. The present document is consistent with this principle. Our intent is to present objective criteria for diagnosis and staging AD, incorporating recent advances in biomarkers, to serve as a bridge between research and clinical care. These criteria are not intended to provide step-by-step clinical practice guidelines for clinical workflow or specific treatment protocols, but rather serve as general principles to inform diagnosis and staging of AD that reflect current science. HIGHLIGHTS: We define Alzheimer's disease (AD) to be a biological process that begins with the appearance of AD neuropathologic change (ADNPC) while people are asymptomatic. Progression of the neuropathologic burden leads to the later appearance and progression of clinical symptoms. Early-changing Core 1 biomarkers (amyloid positron emission tomography [PET], approved cerebrospinal fluid biomarkers, and accurate plasma biomarkers [especially phosphorylated tau 217]) map onto either the amyloid beta or AD tauopathy pathway; however, these reflect the presence of ADNPC more generally (i.e., both neuritic plaques and tangles). An abnormal Core 1 biomarker result is sufficient to establish a diagnosis of AD and to inform clinical decision making throughout the disease continuum. Later-changing Core 2 biomarkers (biofluid and tau PET) can provide prognostic information, and when abnormal, will increase confidence that AD is contributing to symptoms. An integrated biological and clinical staging scheme is described that accommodates the fact that common copathologies, cognitive reserve, and resistance may modify relationships between clinical and biological AD stages.
Collapse
Affiliation(s)
| | - J. Scott Andrews
- Global Evidence & OutcomesTakeda Pharmaceuticals Company LimitedCambridgeMassachusettsUSA
| | - Thomas G. Beach
- Civin Laboratory for NeuropathologyBanner Sun Health Research InstituteSun CityArizonaUSA
| | - Teresa Buracchio
- Office of NeuroscienceU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Ana Graf
- NovartisNeuroscience Global Drug DevelopmentBaselSwitzerland
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Faculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University Hospital, MalmöLundSweden
| | - Carole Ho
- DevelopmentDenali TherapeuticsSouth San FranciscoCaliforniaUSA
| | - William Jagust
- School of Public Health and Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Eric McDade
- Department of NeurologyWashington University St. Louis School of MedicineSt. LouisMissouriUSA
| | - Jose Luis Molinuevo
- Department of Global Clinical Development H. Lundbeck A/SExperimental MedicineCopenhagenDenmark
| | - Ozioma C. Okonkwo
- Department of Medicine, Division of Geriatrics and GerontologyUniversity of Wisconsin School of MedicineMadisonWisconsinUSA
| | - Luca Pani
- University of MiamiMiller School of MedicineMiamiFloridaUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research Institute (ATRI)Keck School of Medicine at the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Philip Scheltens
- Amsterdam University Medical Center (Emeritus)NeurologyAmsterdamthe Netherlands
| | - Eric Siemers
- Clinical ResearchAcumen PharmaceuticalsZionsvilleIndianaUSA
| | - Heather M. Snyder
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| | - Reisa Sperling
- Department of Neurology, Brigham and Women's HospitalMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Charlotte E. Teunissen
- Department of Laboratory MedicineAmsterdam UMC, Neurochemistry LaboratoryAmsterdamthe Netherlands
| | - Maria C. Carrillo
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
47
|
Kobayashi E. The Crossroads Between Alzheimer's Disease Pathophysiology and Epilepsy. Epilepsy Curr 2024; 24:245-247. [PMID: 39309046 PMCID: PMC11412408 DOI: 10.1177/15357597241256616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Association of Plasma Aβ42/Aβ40 Ratio and Late-Onset Epilepsy: Results From the Atherosclerosis Risk in Communities Study Johnson EL, Sullivan KJ, Schneider ALC, Simino J, Mosley TH, Kucharska-Newton A, Knopman DS, Gottesman RF. Neurology . 2023 Sep 26;101(13):e1319-e1327. doi:10.1212/WNL.0000000000207635 . Epub 2023 Aug 4. PMID: 37541842 Background and Objectives: The objective of this study was to determine the relationship between plasma β-amyloid (Aβ), specifically the ratio of 2 Aβ peptides (the Aβ42/Aβ40 ratio, which correlates with increased accumulation of Aβ in the central nervous system [CNS]), and late-onset epilepsy (LOE). Methods: We used Medicare fee-for-service claims codes from 1991 to 2018 to identify cases of LOE among 1424 Black and White men and women enrolled in the Atherosclerosis Risk in Communities (ARIC) study cohort. The Aβ42/Aβ40 ratio was calculated from plasma samples collected from ARIC participants from 1993 to 1995 (age 50-71 years) and 2011 to 2013 (age 67-90 years). We used survival analysis accounting for the competing risk of death to determine the relationship between late-life plasma Aβ42/Aβ40, and its change from midlife to late life, and the subsequent development of epilepsy. We adjusted for demographics, the apolipoprotein e4 genotype, and comorbidities, including stroke, dementia, and head injury. A low plasma ratio of 2 Aβ peptides, the Aβ42/Aβ40 ratio, correlates with low CSF Aβ42/Aβ40 and with increased accumulation of Aβ in the CNS. Results: A decrease in plasma Aβ42/Aβ40 ratio from midlife to late life, but not an isolated measurement of Aβ42/Aβ40, was associated with the development of epilepsy in later life. For every 50% reduction in Aβ42/Aβ40, there was a 2-fold increase in the risk of epilepsy (adjusted subhazard ratio 2.30, 95% CI: 1.27-4.17). Discussion: A reduction in plasma Aβ42/Aβ40 is associated with an increased risk of subsequent epilepsy. Our observations provide a further validation of the link between Aβ, hyperexcitable states, and LOE. Similar Brain Proteomic Signatures in Alzheimer's Disease and Epilepsy Leitner D, Pires G, Kavanagh T, Kanshin E, Askenazi M, Ueberheide B, Devinsky O, Wisniewski T, Drummond E. Acta Neuropathol . 2024 Jan 30;147(1):27. doi:10.1007/s00401-024-02683-4 . PMID: 38289539 The prevalence of epilepsy is increased among Alzheimer's disease (AD) patients and cognitive impairment is common among people with epilepsy. Epilepsy and AD are linked but the shared pathophysiological changes remain poorly defined. We aim to identify protein differences associated with epilepsy and AD using published proteomics datasets. We observed a highly significant overlap in protein differences in epilepsy and AD: 89% (689/777) of proteins altered in the hippocampus of epilepsy patients were significantly altered in advanced AD. Of the proteins altered in both epilepsy and AD, 340 were altered in the same direction, while 216 proteins were altered in the opposite direction. Synapse and mitochondrial proteins were markedly decreased in epilepsy and AD, suggesting common disease mechanisms. In contrast, ribosome proteins were increased in epilepsy but decreased in AD. Notably, many of the proteins altered in epilepsy interact with tau or are regulated by tau expression. This suggests that tau likely mediates common protein changes in epilepsy and AD. Immunohistochemistry for Aβ and multiple phosphorylated tau species (pTau396/404, pTau217, and pTau231) showed a trend for increased intraneuronal pTau217 and pTau231 but no phosphorylated tau aggregates or amyloid plaques in epilepsy hippocampal sections. Our results provide insights into common mechanisms in epilepsy and AD and highlight the potential role of tau in mediating common pathological protein changes in epilepsy and AD.
Collapse
Affiliation(s)
- Eliane Kobayashi
- Department of Neurology and Neurosurgery
- Montreal Neurological Institute and Hospital
- Faculty of Medicine and Health Sciences
- McGill University
| |
Collapse
|
48
|
Arranz J, Zhu N, Rubio-Guerra S, Rodríguez-Baz Í, Ferrer R, Carmona-Iragui M, Barroeta I, Illán-Gala I, Santos-Santos M, Fortea J, Lleó A, Tondo M, Alcolea D. Diagnostic performance of plasma pTau 217, pTau 181, Aβ 1-42 and Aβ 1-40 in the LUMIPULSE automated platform for the detection of Alzheimer disease. Alzheimers Res Ther 2024; 16:139. [PMID: 38926773 PMCID: PMC11200993 DOI: 10.1186/s13195-024-01513-9] [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: 12/08/2023] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Recently developed blood markers for Alzheimer's disease (AD) detection have high accuracy but usually require ultra-sensitive analytic tools not commonly available in clinical laboratories, and their performance in clinical practice is unknown. METHODS We analyzed plasma samples from 290 consecutive participants that underwent lumbar puncture in routine clinical practice in a specialized memory clinic (66 cognitively unimpaired, 130 participants with mild cognitive impairment, and 94 with dementia). Participants were classified as amyloid positive (A +) or negative (A-) according to CSF Aβ1-42/Aβ1-40 ratio. Plasma pTau217, pTau181, Aβ1-42 and Aβ1-40 were measured in the fully-automated LUMIPULSE platform. We used linear regression to compare plasma biomarkers concentrations between A + and A- groups, evaluated Spearman's correlation between plasma and CSF and performed ROC analyses to assess their diagnostic accuracy to detect brain amyloidosis as determined by CSF Aβ1-42/Aβ1-40 ratio. We analyzed the concordance of pTau217 with CSF amyloidosis. RESULTS Plasma pTau217 and pTau181 concentration were higher in A + than A- while the plasma Aβ1-42/Aβ1-40 ratio was lower in A + compared to A-. pTau181 and the Aβ1-42/Aβ1-40 ratio showed moderate correlation between plasma and CSF (Rho = 0.66 and 0.69, respectively). The areas under the ROC curve to discriminate A + from A- participants were 0.94 (95% CI 0.92-0.97) for pTau217, and 0.88 (95% CI 0.84-0.92) for both pTau181 and Aβ1-42/Aβ1-40. Chronic kidney disease (CKD) was related to increased plasma biomarker concentrations, but ratios were less affected. Plasma pTau217 had the highest fold change (× 3.2) and showed high predictive capability in discriminating A + from A-, having 4-7% misclassification rate. The global accuracy of plasma pTau217 using a two-threshold approach was robust in symptomatic groups, exceeding 90%. CONCLUSION The evaluation of blood biomarkers on an automated platform exhibited high diagnostic accuracy for AD pathophysiology, and pTau217 showed excellent diagnostic accuracy to identify participants with AD in a consecutive sample representing the routine clinical practice in a specialized memory unit.
Collapse
Affiliation(s)
- Javier Arranz
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Department of Neurology, Unidad Alzheimer-Down, IR SANT PAU, Hospital de La Santa Creu I Sant Pau; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nuole Zhu
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sara Rubio-Guerra
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Íñigo Rodríguez-Baz
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Department of Neurology, Unidad Alzheimer-Down, IR SANT PAU, Hospital de La Santa Creu I Sant Pau; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Rosa Ferrer
- Servei de Bioquímica I Biologia Molecular, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, C/Sant Quintí 89, 08041, Barcelona, Spain
| | - María Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Department of Neurology, Unidad Alzheimer-Down, IR SANT PAU, Hospital de La Santa Creu I Sant Pau; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Department of Neurology, Unidad Alzheimer-Down, IR SANT PAU, Hospital de La Santa Creu I Sant Pau; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Miguel Santos-Santos
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Department of Neurology, Unidad Alzheimer-Down, IR SANT PAU, Hospital de La Santa Creu I Sant Pau; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Mireia Tondo
- Servei de Bioquímica I Biologia Molecular, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, C/Sant Quintí 89, 08041, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas, CIBERDEM, Madrid, Spain.
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain.
| |
Collapse
|
49
|
Ossenkoppele R, Salvadó G, Janelidze S, Binette AP, Bali D, Karlsson L, Palmqvist S, Mattsson-Carlgren N, Stomrud E, Therriault J, Rahmouni N, Rosa-Neto P, Coomans EM, van de Giessen E, van der Flier WM, Teunissen CE, Jonaitis EM, Johnson SC, PREVENT-AD Research Group, Villeneuve S, Benzinger TL, Schindler SE, Bateman RJ, Doecke JD, Doré V, Feizpour A, Masters CL, Rowe C, Wiste HJ, Petersen RC, Jack CR, Hansson O. Prediction of future cognitive decline among cognitively unimpaired individuals using measures of soluble phosphorylated tau or tau tangle pathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.12.24308824. [PMID: 38947004 PMCID: PMC11213114 DOI: 10.1101/2024.06.12.24308824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Plasma p-tau217 and Tau-PET are strong prognostic biomarkers in Alzheimer's disease (AD), but their relative performance in predicting future cognitive decline among cognitively unimpaired (CU) individuals is unclear. In this head-to-head comparison study including 9 cohorts and 1534 individuals, we found that plasma p-tau217 and medial temporal lobe Tau-PET signal showed similar associations with cognitive decline on a global cognitive composite test (R2 PET=0.32 vs R2 PLASMA=0.32, pdifference=0.812) and with progression to mild cognitive impairment (Hazard ratio[HR]PET=1.56[1.43-1.70] vs HRPLASMA=1.63[1.50-1.77], pdifference=0.627). Combined plasma and PET models were superior to the single biomarker models (R2=0.36, p<0.01). Furthermore, sequential selection using plasma p-tau217 and then Tau-PET reduced the number of participants required for a clinical trial by 94%, compared to a 75% reduction when using plasma p-tau217 alone. We conclude that plasma p-tau217 and Tau-PET showed similar performance for predicting future cognitive decline in CU individuals, and their sequential use (i.e., plasma p-tau217 followed by Tau-PET in a subset with high plasma p-tau217) is useful for screening in clinical trials in preclinical AD.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Emma M. Coomans
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Neurochemistry Laboratory, Department of Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Erin M. Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | | | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Randall J. Bateman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
| | - James D. Doecke
- Australian eHealth Research Centre, CSIRO, Melbourne, Victoria, Australia
| | - Vincent Doré
- Australian eHealth Research Centre, CSIRO, Melbourne, Victoria, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - Azadeh Feizpour
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
50
|
Sato S, Hatakeyama N, Fujikoshi S, Katayama S, Katagiri H, Sims JR. Donanemab in Japanese Patients with Early Alzheimer's Disease: Subpopulation Analysis of the TRAILBLAZER-ALZ 2 Randomized Trial. Neurol Ther 2024; 13:677-695. [PMID: 38581616 PMCID: PMC11136931 DOI: 10.1007/s40120-024-00604-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/14/2024] [Indexed: 04/08/2024] Open
Abstract
INTRODUCTION Donanemab, a monoclonal antibody directed against an insoluble, modified, N-terminal truncated form of amyloid beta, demonstrated efficacy and safety in patients with early, symptomatic Alzheimer's disease (AD) in the phase 3 TRAILBLAZER-ALZ 2 trial. Here, we report clinical outcomes, biomarkers, and safety results for the Japanese subpopulation. METHODS TRAILBLAZER-ALZ 2 (N = 1736) was conducted in eight countries, including Japan (enrollment June 2020-November 2021; database lock April 2023). Participants (60-85 years) with early, symptomatic AD (mild cognitive impairment/mild dementia), Mini-Mental State Examination score 20-28, and confirmed amyloid and tau pathology were randomized 1:1 (stratified by tau status) to intravenous donanemab (700 mg for three doses, then 1400 mg/dose) or placebo every 4 weeks for 72 weeks. Primary outcome was change from baseline to week 76 in integrated Alzheimer's Disease Rating Scale (iADRS) score. Other outcomes included clinical measures of cognitive and functional impairment, biomarkers, and safety. RESULTS Of 88 Japanese participants (43 placebo, 45 donanemab), 7 in each group discontinued. Least-squares mean (LSM) change from baseline in iADRS score at week 76 was smaller with donanemab than with placebo in the combined (low-medium tau and high tau) and low-medium tau (N = 76) subpopulations (LSM change difference: 4.43 and 3.99, representing 38.8% and 40.2% slowing of disease progression, respectively). Slowing of AD progression with donanemab was also observed for other clinical outcomes. Marked decreases in amyloid plaque and plasma phosphorylated tau 217 were observed; amyloid clearance (< 24.1 Centiloids) was observed in 83.3% of the combined donanemab and 0% of the combined placebo groups. Amyloid-related imaging abnormalities of edema/effusions occurred in ten (22.2%) donanemab-treated participants (one [2.2%] symptomatic) and one (2.3%) placebo-treated participant. CONCLUSIONS The overall efficacy and safety of donanemab in Japanese participants were similar to the global TRAILBLAZER-ALZ 2 population. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT04437511.
Collapse
Affiliation(s)
- Shoichiro Sato
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K., 5-1-28, Isogamidori, Chuo-Ku, Kobe, 651-0086, Japan.
| | - Naohisa Hatakeyama
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K., 5-1-28, Isogamidori, Chuo-Ku, Kobe, 651-0086, Japan
| | - Shinji Fujikoshi
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K., 5-1-28, Isogamidori, Chuo-Ku, Kobe, 651-0086, Japan
| | | | - Hideaki Katagiri
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K., 5-1-28, Isogamidori, Chuo-Ku, Kobe, 651-0086, Japan
| | - John R Sims
- Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|