1
|
Wang Z, Chen Y, Gong K, Zhao B, Ning Y, Chen M, Li Y, Ali M, Timsina J, Liu M, Cruchaga C, Jia J. Cerebrospinal fluid proteomics identification of biomarkers for amyloid and tau PET stages. Cell Rep Med 2025; 6:102031. [PMID: 40118053 DOI: 10.1016/j.xcrm.2025.102031] [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: 09/24/2024] [Revised: 01/15/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025]
Abstract
Accurate staging of Alzheimer's disease (AD) pathology is crucial for therapeutic trials and prognosis, but existing fluid biomarkers lack specificity, especially for assessing tau deposition severity, in amyloid-beta (Aβ)-positive patients. We analyze cerebrospinal fluid (CSF) samples from 136 participants in the Alzheimer's Disease Neuroimaging Initiative using more than 6,000 proteins. We apply machine learning to predict AD pathological stages defined by amyloid and tau positron emission tomography (PET). We identify two distinct protein panels: 16 proteins, including neurofilament heavy chain (NEFH) and SPARC-related modular calcium-binding protein 1 (SMOC1), that distinguished Aβ-negative/tau-negative (A-T-) from A+ individuals and nine proteins, such as HCLS1-associated protein X-1 (HAX1) and glucose-6-phosphate isomerase (GPI), that differentiated A+T+ from A+T- stages. These signatures outperform the established CSF biomarkers (area under the curve [AUC]: 0.92 versus 0.67-0.70) and accurately predicted disease progression over a decade. The findings are validated in both internal and external cohorts. These results underscore the potential of proteomic-based signatures to refine AD diagnostic criteria and improve patient stratification in clinical trials.
Collapse
Affiliation(s)
- Zhibo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Yuhan Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou 075000, China
| | - Katherine Gong
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Bote Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Yuye Ning
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Meilin Chen
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurologic Diseases, Washington University, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA.
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, P.R.China; Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing 100053, P.R.China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing 100053, P.R.China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100053, P.R.China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, P.R.China.
| |
Collapse
|
2
|
Cummings JL, Teunissen CE, Fiske BK, Le Ber I, Wildsmith KR, Schöll M, Dunn B, Scheltens P. Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 2025:10.1038/s41573-025-01165-w. [PMID: 40185982 DOI: 10.1038/s41573-025-01165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders are characterized by complex neurobiological changes that are reflected in biomarker alterations detectable in blood, cerebrospinal fluid (CSF) and with brain imaging. As accessible proxies for processes that are difficult to measure, biomarkers are tools that hold increasingly important roles in drug development and clinical trial decision making. In the past few years, biomarkers have been the basis for accelerated approval of new therapies for Alzheimer disease and amyotrophic lateral sclerosis as surrogate end points reasonably likely to predict clinical benefit.Blood-based biomarkers are emerging for Alzheimer disease and other neurodegenerative disorders (for example, Parkinson disease, frontotemporal dementia), and some biomarkers may be informative across multiple disease states. Collection of CSF provides access to biomarkers not available in plasma, including markers of synaptic dysfunction and neuroinflammation. Molecular imaging is identifying an increasing array of targets, including amyloid plaques, neurofibrillary tangles, inflammation, mitochondrial dysfunction and synaptic density. In this Review, we consider how biomarkers can be implemented in clinical trials depending on their context of use, including providing information on disease risk and/or susceptibility, diagnosis, prognosis, pharmacodynamic outcomes, monitoring, prediction of response to therapy and safety. Informed choice of increasingly available biomarkers and rational deployment in clinical trials support drug development decision making and de-risk the drug development process for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Göteborg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Philip Scheltens
- Alzheimer's Center Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
- EQT Group, Dementia Fund, Stockholm, Sweden
| |
Collapse
|
3
|
Schöll M, Vrillon A, Ikeuchi T, Quevenco FC, Iaccarino L, Vasileva-Metodiev SZ, Burnham SC, Hendrix J, Epelbaum S, Zetterberg H, Palmqvist S. Cutting through the noise: A narrative review of Alzheimer's disease plasma biomarkers for routine clinical use. J Prev Alzheimers Dis 2025; 12:100056. [PMID: 39814656 DOI: 10.1016/j.tjpad.2024.100056] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
As novel, anti-amyloid therapies have become more widely available, access to timely and accurate diagnosis has become integral to ensuring optimal treatment of patients with early-stage Alzheimer's disease (AD). Plasma biomarkers are a promising tool for identifying AD pathology; however, several technical and clinical factors need to be considered prior to their implementation in routine clinical use. Given the rapid pace of advancements in the field and the wide array of available biomarkers and tests, this review aims to summarize these considerations, evaluate available platforms, and discuss the steps needed to bring plasma biomarker testing to the clinic. We focus on plasma phosphorylated(p)-tau, specifically plasma p-tau217, as a robust candidate across both primary and secondary care settings. Despite the high performance and robustness demonstrated in research, plasma p-tau217, like all plasma biomarkers, can be affected by analytical and pre-analytical variability as well as patient comorbidities, sex, ethnicity, and race. This review also discusses the advantages of the two-point cut-off approach to mitigating these factors, and the challenges raised by the resulting intermediate range measurements, where clinical guidance is still unclear. Further validation of plasma p-tau217 in heterogeneous, real-world cohorts will help to increase confidence in testing and support establishing a standardized approach. Plasma biomarkers are poised to become a more affordable and less invasive alternative to PET and CSF testing. However, understanding the factors that impact plasma biomarker measurement and interpretation is critical prior to their implementation in routine clinical use.
Collapse
Affiliation(s)
- M Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK; Department of Neuropsychiatry, Sahlgrenska University Hospital, Mölndal, Sweden
| | - A Vrillon
- French Institute of Health and Medical Research (Inserm), Paris, France
| | - T Ikeuchi
- Niigata University Brain Research Institute, Niigata, Japan
| | - F C Quevenco
- Eli Lilly and Company, Indianapolis, IN, United States
| | - L Iaccarino
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | - S C Burnham
- Eli Lilly and Company, Indianapolis, IN, United States
| | - J Hendrix
- Eli Lilly and Company, Indianapolis, IN, United States
| | - S Epelbaum
- Eli Lilly and Company, Indianapolis, IN, United States
| | - H Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - S Palmqvist
- Clinical Memory Research Unit, Clinical Sciences in Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Sweden.
| |
Collapse
|
4
|
Zeiss CJ, Huttner A, Nairn AC, Arnsten A, Datta D, Strittmatter SM, Wyk BV, Duque A. The neuropathologic basis for translational biomarker development in the macaque model of late-onset Alzheimer's disease. J Alzheimers Dis 2025; 104:1243-1258. [PMID: 40095666 DOI: 10.1177/13872877251323787] [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] [Indexed: 03/19/2025]
Abstract
BackgroundAccurate placement of the macaque within the Alzheimer's disease (AD) research framework is essential to discover early-stage predictive biomarkers.ObjectiveTo assess utility of the aging macaque in advancing translational biomarker development for preclinical AD, we evaluated relative signal strength of comparable neuropathologic phenomena in macaques and patients.MethodsWe compared pathology in patient and macaque formalin-fixed paraffin embedded (FFPE) tissues using identical criteria. We quantified expression of amyloid-β (Aβ), pTau, and inflammatory and senescence markers across species. Distribution of AD-relevant markers were compared in FFPE and perfused frozen macaque brain to assess expression of labile proteins that could inform in-life fluid biomarkers.ResultsAβ pathology in macaques closely approximated patient pathology. Complex plaque composition in macaques implied significant disruption of synaptic connectivity. In FFPE tissue, pretangle pTau immunoreactivity placed the macaque in Braak Stage 1b. In perfused frozen tissue, soluble pTau distribution approximated Braak Stage III-IV. In macaque, Aβ, pTau, and acetylcholinesterase labeling co-localized to AD-vulnerable circuits. Significant association of glial fibrillary acidic protein with Aβ occurred in humans only. The senescence marker p16 correlated positively with pTau expression and negatively with Aβ in patients only. Macaques lacked neuropathologic co-morbidities.ConclusionsAD-relevant neuropathologic signals in the macaque support biomarker discovery in the areas of Aβ plaque evolution and associated synaptic disruption as well as early-stage tau phosphorylation. Relative protection from accumulation of senescence markers, fibrillar tau and neuropathologic co-morbidities in macaque implicate species difference in rates of biological brain aging. We provide over 4000 digital slides for further study.
Collapse
Affiliation(s)
- Caroline J Zeiss
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Amy Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen M Strittmatter
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
5
|
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] [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
|
6
|
Borelli WV, Ferreira PCL, Brum WS, Ferrari-Souza JP, Carello-Collar G, Holz M, Tizeli V, Strelow MZ, Formoso C, Fagundes Chaves ML, Rocha A, Aguzzoli CS, Rohden F, Souza DG, Schumacher Schuh AF, Povala G, Bellaver B, Rosa-Neto P, Castilhos RM, Pascoal TA, Zimmer ER. Diagnostic performance of Alzheimer's disease blood biomarkers in a Brazilian cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.23.24319116. [PMID: 40271009 PMCID: PMC12016014 DOI: 10.1101/2025.02.23.24319116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Blood-based biomarkers (BBMs) have emerged as promising tools to enhance Alzheimer's disease (AD) diagnosis. Despite two-thirds of dementia cases occurring in the Global South, research on BBMs has predominantly focused on populations from the Global North. This geographical disparity hinders our understanding of BBM performance in diverse populations. To address this, we evaluated the diagnostic properties of AD BBMs in a real-world memory clinic from Brazil, one of the largest countries in the Global South. We measured blood and cerebrospinal fluid (CSF) biomarkers - amyloid-β (Aβ)40, Aβ42, phosphorylated tau (p-tau) 217, neurofilament light (NfL) chain, and glial fibrillary acidic protein (GFAP) - in 59 individuals. Sample comprised 20 cognitively unimpaired (CU) individuals, 22 with AD dementia, and 17 with vascular dementia (VaD). We compared BBM levels across diagnostic groups and assessed their discriminative ability for AD. Notably, individuals with VaD and AD had lower educational levels (6.8±3.0) compared to CU individuals (61.4±6.6). Among the BBMs tested, plasma p-tau217 demonstrated the best performance, exhibiting high accuracy in differentiating CU from AD (AUC 0.96) and Aβ pathology (AUC 0.98). However, the ability of AD BBMs to distinguish between AD and VaD was lower than expected (AUC from 0.52 to 0.79), particularly when compared to studies from the Global North. Our findings highlight the potential utility of BBMs for AD diagnosis in real-world settings within the Global South. However, they also underscore the need for proper implementation and validation of these biomarkers within these populations to ensure accurate and reliable results.
Collapse
|
7
|
Olvera‐Rojas M, Sewell KR, Karikari TK, Huang H, Oberlin LE, Zeng X, Morris JK, Collins AM, Drake JA, Sutton BP, Kramer AF, Hillman CH, Vidoni ED, Burns JM, Kamboh MI, McAuley E, Marsland AL, Chen Y, Lafferty TK, Sehrawat A, Jakicic JM, Wan L, Kang C, Erickson KI. Influence of medical conditions on the diagnostic accuracy of plasma p-tau217 and p-tau217/Aβ42. Alzheimers Dement 2025; 21:e14430. [PMID: 39641408 PMCID: PMC11848339 DOI: 10.1002/alz.14430] [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: 09/09/2024] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Blood-based biomarkers (BBMs) can enable early detection of brain amyloid beta (Aβ) pathology in cognitively unimpaired individuals. However, the extent to which common medical conditions affect biomarker performance remains unclear. METHODS Participants (n = 348) included individuals without cognitive impairment. We studied how brain Aβ associated with BBMs (Aβ42/40, phosphorylated tau [p-tau] 181 and 217, p-tau217/Aβ42, glial fibrillary acidic protein [GFAP], and neurofilament light [NfL]) and optimal BBM thresholds for predicting brain Aβ positivity and whether they are obscured by the presence of common medical conditions. RESULTS Plasma Aβ42/40, p-tau181, p-tau217, and GFAP, but not NfL, were significantly associated with brain Aβ. P-tau217/Aβ42 showed the best discriminative performance (area under the curve: 0.91). The strength of p-tau217-brain Aβ associations were obscured by diabetes and cardiovascular conditions. DISCUSSION These results suggest BBMs may help detect early Aβ pathology but suggest caution in their use due to common medical conditions that could affect accuracy. HIGHLIGHTS Plasma Aβ42/40, p-tau181, p-tau217, and GFAP but not NfL showed significant associations with brain Aβ. BBMs were more strongly associated with the level of brain Aβ in those without diabetes and cardiovascular conditions. P-tau217/Aβ42 showed the best performance (AUC = 0.91) in discriminating Aβ presence with an optimal cut-off of >1.2, followed by p-tau217 at >0.46 pg/mL, with performance slightly improving when excluding participants with cardiovascular conditions.
Collapse
Affiliation(s)
- Marcos Olvera‐Rojas
- Department of Physical Education and Sports, Faculty of Sport SciencesSport and Health University Research Institute (iMUDS), University of GranadaGranadaSpain
| | - Kelsey R. Sewell
- AdventHealth Research InstituteOrlandoFloridaUSA
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
| | - Thomas K. Karikari
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Lauren E. Oberlin
- AdventHealth Research InstituteOrlandoFloridaUSA
- Department of PsychiatryWeill Cornell MedicineNew YorkNew YorkUSA
| | - Xuemei Zeng
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jill K. Morris
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | | | - Jermon A. Drake
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bradley P. Sutton
- The Grainger College of Engineering, Bioengineering DepartmentUniversity of IllinoisChampaignIllinoisUSA
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Arthur F. Kramer
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Center for Cognitive and Brain HealthNortheastern UniversityBostonMassachusettsUSA
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
| | - Charles H. Hillman
- Center for Cognitive and Brain HealthNortheastern UniversityBostonMassachusettsUSA
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
- Department of Physical Therapy, Movement, & Rehabilitation SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Eric D. Vidoni
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Jeffrey M. Burns
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - M. Ilyas Kamboh
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
- University of Pittsburgh Department of EpidemiologyPittsburghPennsylvaniaUSA
- University of Pittsburgh Department of Human GeneticsPittsburghPennsylvaniaUSA
| | - Edward McAuley
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Anna L. Marsland
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Yijun Chen
- Department of Chemistry, Chevron Science CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Tara K. Lafferty
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Anuradha Sehrawat
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - John M. Jakicic
- Department of Internal Medicine, Division of Physical Activity and Weight ManagementUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Lu Wan
- AdventHealth Research InstituteOrlandoFloridaUSA
| | - Chaeryon Kang
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kirk I. Erickson
- AdventHealth Research InstituteOrlandoFloridaUSA
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
8
|
Chen Y, Albert AL, Sehrawat A, Farinas M, Lopez OL, Zeng X, Cohen AD, Karikari TK. Equivalence of plasma and serum for clinical measurement of p-tau217: comparative analyses of four blood-based assays. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.26.24319657. [PMID: 39763537 PMCID: PMC11703320 DOI: 10.1101/2024.12.26.24319657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Background Phosphorylated tau (p-tau) 217 is a promising blood biomarker for Alzheimer's disease (AD). However, most p-tau217 assays have been validated solely in ethylenediaminetetraacetic acid (EDTA) plasma, leaving the clinical applicability of serum p-tau217 largely unexplored despite serum being a preferred matrix in many clinical laboratories. To address this gap, we compared p-tau217 concentrations and diagnostic performances in matched plasma and serum samples using four research-use-only assays, including three from commercial sources i.e., Lumipulse, ALZpath, NULISA, and one from University of Pittsburgh. Methods Paired plasma and serum samples were processed from the same venipuncture collection and assessed with the four p-tau217 assays following manufacturer-recommended procedures in two research cohorts (N=84). Results Plasma and serum p-tau217 levels varied across assays; the ALZpath, Pittsburgh, and NULISA methods showed significantly lower p-tau217 levels in serum compared with plasma (p<0.0001), while Lumipulse showed higher or non-significant differences in serum. Yet, strong correlations (rho >0.8) were observed between plasma and serum p-tau217 pairs. Both plasma and serum p-tau217 demonstrated strong classification accuracies to differentiate clinical AD from normal controls, with high AUC (up to 0.963) for all methods. The exception was the Pittsburgh assay, where plasma p-tau217 had superior AUC than serum p-tau217 (plasma: 0.912, serum: 0.844). The rest of the assays had equivalent accuracies in both matrices. Conclusions Serum p-tau217 performs equivalently as plasma p-tau217 for most assessed assays. Serum can therefore be used in place of plasma for p-tau217 assessment for research and clinical purposes.
Collapse
Affiliation(s)
- Yijun Chen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ally L Albert
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Marissa Farinas
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Oscar L Lopez
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ann D Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Thomas K Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
9
|
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: 0] [Impact Index Per Article: 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
|
10
|
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
|
11
|
Mitolo M, Lombardi G, Manca R, Nacmias B, Venneri A. Association between blood-based protein biomarkers and brain MRI in the Alzheimer's disease continuum: a systematic review. J Neurol 2024; 271:7120-7140. [PMID: 39264441 PMCID: PMC11560990 DOI: 10.1007/s00415-024-12674-w] [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: 07/19/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
Blood-based biomarkers (BBM) are becoming easily detectable tools to reveal pathological changes in Alzheimer's disease (AD). A comprehensive and up-to-date overview of the association between BBM and brain MRI parameters is not available. This systematic review aimed to summarize the literature on the associations between the main BBM and MRI markers across the clinical AD continuum. A systematic literature search was carried out on PubMed and Web of Science and a total of 33 articles were included. Hippocampal volume was positively correlated with Aβ42 and Aβ42/Aβ40 and negatively with Aβ40 plasma levels. P-tau181 and p-tau217 concentrations were negatively correlated with temporal grey matter volume and cortical thickness. NfL levels were negatively correlated with white matter microstructural integrity, whereas GFAP levels were positively correlated with myo-inositol values in the posterior cingulate cortex/precuneus. These findings highlight consistent associations between various BBM and brain MRI markers even in the pre-clinical and prodromal stages of AD. This suggests a possible advantage in combining multiple AD-related markers to improve accuracy of early diagnosis, prognosis, progression monitoring and treatment response.
Collapse
Affiliation(s)
- Micaela Mitolo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Gemma Lombardi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi Onlus, Florence, Italy
| | - Riccardo Manca
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
- Department of Life Sciences, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK.
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi Onlus, Florence, Italy
| | - Annalena Venneri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Department of Life Sciences, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| |
Collapse
|
12
|
Lin X, Chen C, Chen J, Zhu C, Zhang J, Su R, Chen S, Weng S, Chang X, Lin S, Chen Y, Li J, Lin L, Zhou J, Guo Z, Yu G, Shao W, Hu H, Wu S, Zhang Q, Li H, Zheng F. Long Noncoding RNA NR_030777 Alleviates Cobalt Nanoparticles-Induced Neurodegenerative Damage by Promoting Autophagosome-Lysosome Fusion. ACS NANO 2024; 18:24872-24897. [PMID: 39197041 PMCID: PMC11394346 DOI: 10.1021/acsnano.4c05249] [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: 04/21/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 08/30/2024]
Abstract
Potential exposure to cobalt nanoparticles (CoNPs) occurs in various fields, including hard alloy industrial production, the increasing use of new energy lithium-ion batteries, and millions of patients with metal-on-metal joint prostheses. Evidence from human, animal, and in vitro experiments suggests a close relationship between CoNPs and neurotoxicity. However, a systematic assessment of central nervous system (CNS) impairment due to CoNPs exposure and the underlying molecular mechanisms is lacking. In this study, we found that CoNPs induced neurodegenerative damage both in vivo and in vitro, including cognitive impairment, β-amyloid deposition and Tau hyperphosphorylation. CoNPs promoted the formation of autophagosomes and impeding autophagosomal-lysosomal fusion in vivo and in vitro, leading to toxic protein accumulation. Moreover, CoNPs exposure reduced the level of transcription factor EB (TFEB) and the abundance of lysosome, causing a blockage in autophagosomal-lysosomal fusion. Interestingly, overexpression of long noncoding RNA NR_030777 mitigated CoNPs-induced neurodegenerative damage in both in vivo and in vitro models. Fluorescence in situ hybridization assay revealed that NR_030777 directly binds and stabilizes TFEB mRNA, alleviating the blockage of autophagosomal-lysosomal fusion and ultimately restoring neurodegeneration induced by CoNPs in vivo and in vitro. In summary, our study demonstrates that autophagic dysfunction is the main toxic mechanism of neurodegeneration upon CoNPs exposure and NR_030777 plays a crucial role in CoNPs-induced autophagic dysfunction. Additionally, the proposed adverse outcome pathway contributes to a better understanding of CNS toxicity assessment of CoNPs.
Collapse
Affiliation(s)
- Xinpei Lin
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Cheng Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jinxiang Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Canlin Zhu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jiajun Zhang
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Ruiqi Su
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shujia Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shucan Weng
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Xiangyu Chang
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Shengsong Lin
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Yilong Chen
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Jiamei Li
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Ling Lin
- Public
Technology Service Center, Fujian Medical
University, Fuzhou, Fujian Province 350122, China
| | - Jinfu Zhou
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- Medical
Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health
Hospital College of Clinical Medicine for Obstetrics & Gynecology
and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province 350001, China
| | - Zhenkun Guo
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Guangxia Yu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Wenya Shao
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Hong Hu
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Siying Wu
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- Department
of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Qunwei Zhang
- Department
of Epidemiology and Population Health, School of Public Health and
Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, Kentucky 40292, United States
| | - Huangyuan Li
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| | - Fuli Zheng
- Department
of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
- The
Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province 350122, China
| |
Collapse
|
13
|
Chen Y, Zeng X, Diaz JL, Sehrawat A, Lafferty TK, Boslett JJ, Klunk WE, Pascoal TA, Villemagne VL, Cohen AD, Lopez O, Yates NA, Karikari TK. Effect of blood collection tube containing protease inhibitors on the pre-analytical stability of Alzheimer's disease plasma biomarkers. J Neurochem 2024; 168:2736-2750. [PMID: 38814273 PMCID: PMC11449657 DOI: 10.1111/jnc.16130] [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: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/31/2024]
Abstract
The reliability of plasma biomarkers of Alzheimer's disease (AD) can be compromised by protease-induced degradation. This can limit the feasibility of conducting plasma biomarker studies in environments that lack the capacity for immediate processing and appropriate storage of blood samples. We hypothesized that blood collection tube supplementation with protease inhibitors can improve the stability of plasma biomarkers at room temperatures (RT). In this study, we conducted a comparative analysis of blood biomarker stability in traditional ethylenediaminetetraacetic acid (EDTA) tubes versus BD™ P100 collection tubes, the latter being coated with a protease inhibitor cocktail. The stability of six plasma AD biomarkers was evaluated over time under RT conditions. We evaluated three experimental approaches. In Approach 1, pooled plasma samples underwent storage at RT for up to 96 h. In Approach 2, plasma samples isolated upfront from whole blood collected into EDTA or P100 tubes were stored at RT for 0 h or 24 h before biomarker measurements. In Approach 3, whole blood samples were collected into paired EDTA and P100 tubes, followed by storage at RT for 0 h or 24 h before isolating the plasma for analyses. Biomarkers were measured with Single Molecule Array (Simoa) and immunoprecipitation-mass spectrometry (IP-MS) assays. Both the IP-MS and Simoa methods revealed that the use of P100 tubes significantly improves the stability of Aβ42 and Aβ40 across all approaches. However, the Aβ42/Aβ40 ratio levels were significantly stabilized only in the IP-MS assay in Approach 3. No significant differences were observed in the levels of plasma p-tau181, GFAP, and NfL for samples collected using either tube type in any of the approaches. Supplementation of blood collection tubes with protease inhibitors could reduce the protease-induced degradation of plasma Aβ42 and Aβ40, and the Aβ42/40 ratio for the IP-MS assay. These findings have crucial implications for preanalytical procedures, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Yijun Chen
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jihui L. Diaz
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Tara K. Lafferty
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - James J. Boslett
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - William E. Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Annie D. Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Oscar Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Nathan A. Yates
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
14
|
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: 0] [Impact Index Per Article: 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
|
15
|
Zhang X, Wang J, Zhang Z, Ye K. Tau in neurodegenerative diseases: molecular mechanisms, biomarkers, and therapeutic strategies. Transl Neurodegener 2024; 13:40. [PMID: 39107835 PMCID: PMC11302116 DOI: 10.1186/s40035-024-00429-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/05/2024] [Indexed: 09/14/2024] Open
Abstract
The deposition of abnormal tau protein is characteristic of Alzheimer's disease (AD) and a class of neurodegenerative diseases called tauopathies. Physiologically, tau maintains an intrinsically disordered structure and plays diverse roles in neurons. Pathologically, tau undergoes abnormal post-translational modifications and forms oligomers or fibrous aggregates in tauopathies. In this review, we briefly introduce several tauopathies and discuss the mechanisms mediating tau aggregation and propagation. We also describe the toxicity of tau pathology. Finally, we explore the early diagnostic biomarkers and treatments targeting tau. Although some encouraging results have been achieved in animal experiments and preclinical studies, there is still no cure for tauopathies. More in-depth basic and clinical research on the pathogenesis of tauopathies is necessary.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiangyu Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
16
|
Sintini I, Singh NA, Li D, Mielke MM, Machulda MM, Schwarz CG, Senjem ML, Jack CR, Lowe VJ, Graff‐Radford J, Josephs KA, Whitwell JL. Plasma glial fibrillary acidic protein in the visual and language variants of Alzheimer's disease. Alzheimers Dement 2024; 20:3679-3686. [PMID: 38528318 PMCID: PMC11095421 DOI: 10.1002/alz.13713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 03/27/2024]
Abstract
INTRODUCTION Glial fibrillary acidic protein (GFAP) in plasma is a proxy for astrocytic activity and is elevated in amyloid-β (Aβ)-positive individuals, making GFAP a potential blood-based biomarker for Alzheimer's disease (AD). METHODS We assessed plasma GFAP in 72 Aβ-positive participants diagnosed with the visual or language variant of AD who underwent Aβ- and tau-PET. Fifty-nine participants had follow-up imaging. Linear regression was applied on GFAP and imaging quantities. RESULTS GFAP did not correlate with Aβ- or tau-PET cross-sectionally. There was a limited positive correlation between GFAP and rates of tau accumulation, particularly in the language variant of AD, although associations were weaker after removing one outlier patient with the highest GFAP level. DISCUSSION Among Aβ-positive AD participants with atypical presentations, plasma GFAP did not correlate with levels of AD pathology on PET, suggesting that the associations between GFAP and AD pathology might plateau during the advanced phase of the disease.
Collapse
Affiliation(s)
- Irene Sintini
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | | | - Danni Li
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Michelle M. Mielke
- Department of Epidemiology and PreventionWake Forest UniversityWinston‐SalemNorth CarolinaUSA
| | - Mary M. Machulda
- Department of Psychiatry and PsychologyMayo ClinicRochesterMinnesotaUSA
| | | | | | | | - Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | | | | | | |
Collapse
|
17
|
Chen Y, Zeng X, Lee J, Sehrawat A, Lafferty TK, Boslett JJ, Klunk WE, Pascoal TA, Villemagne VL, Cohen AD, Lopez O, Yates NA, Karikari TK. Effect of blood collection tube containing protease inhibitors on the pre-analytical stability of Alzheimer's disease plasma biomarkers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.05.24303504. [PMID: 38496591 PMCID: PMC10942510 DOI: 10.1101/2024.03.05.24303504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
INTRODUCTION The reliability of plasma Alzheimer's disease (AD) biomarkers can be compromised by protease-induced degradation. This limits the feasibility of conducting plasma biomarker studies in environments that lack the capacity for immediate processing and appropriate storage of blood samples. We hypothesized that blood collection tube supplementation with protease inhibitors can improve the stability of plasma biomarkers at room temperatures (RT). This study conducted a comparative analysis of blood biomarker stability in traditional ethylenediaminetetraacetic acid (EDTA) tubes versus BD™ P100 collection tubes, the latter being coated with a protease inhibitor cocktail. The stability of six plasma AD biomarkers was evaluated over time under RT conditions. METHODS We evaluated three experimental approaches. In Approach 1, pooled plasma samples underwent storage at RT for up to 96 hours. In Approach 2, plasma samples isolated upfront from whole blood collected into EDTA or P100 tubes were stored at RT for 0h or 24h before biomarker measurements. In Approach 3, whole blood samples were collected into paired EDTA or P100 tubes, followed by storage at RT for 0h or 24h before isolating the plasma for analyses. Biomarkers were measured with Single Molecule Array (Simoa) and immunoprecipitation-mass spectrometry (IP-MS) assays. RESULTS Both the IP-MS and Simoa methods revealed that the use of P100 tubes significantly improved the stability of Aβ42 and Aβ40 across all approaches. Additionally, the Aβ42/Aβ40 ratio levels were significantly stabilized only in the IP-MS assay in Approach 3. No significant differences were observed in the levels of plasma p-tau181, GFAP, and NfL for samples collected using either tube type in any of the approaches. CONCLUSION Supplementation of blood collection tubes with protease inhibitors could reduce the protease-induced degradation of plasma Aβ42 and Aβ40, and the Aβ ratio for IP-MS assay. This has crucial implications for preanalytical procedures, particularly in resource-limited settings.
Collapse
|
18
|
Singh N, Sharma S, Ghosh KK, Gupta B, Kuca K. Prominent Perspective on Existing Biological Hallmarks of Alzheimer's Disease. Curr Top Med Chem 2024; 24:1120-1133. [PMID: 38591203 DOI: 10.2174/0115680266292514240404040341] [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: 11/12/2023] [Revised: 02/24/2024] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
Biomarkers are the most significant diagnosis tools tending towards unique approaches and solutions for the prevention and cure of Alzheimer's Disease (AD). The current report provides a clear perception of the concept of various biomarkers and their prominent features through analysis to provide a possible solution for the inhibition of events in AD. Scientists around the world truly believe that crucial hallmarks can serve as critical tools in the early diagnosis, cure, and prevention, as well as the future of medicine. The awareness and understanding of such biomarkers would provide solutions to the puzzled mechanism of this neuronal disorder. Some of the argued biomarkers in the present article are still in an experimental phase as they need to undergo specific clinical trials before they can be considered for treatment.
Collapse
Affiliation(s)
- Namrata Singh
- Department of Engineering Science, Ramrao Adik Institute of Technology, DY Patil University, Navi Mumbai, 400706, India
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Srishti Sharma
- School of Studies in Chemistry, Pt. Ravishankar Shukla University, Raipur, 492010 (C.G.), India
| | - Kallol K Ghosh
- School of Studies in Chemistry, Pt. Ravishankar Shukla University, Raipur, 492010 (C.G.), India
| | - Bhanushree Gupta
- Centre of Basic Sciences, Pt. Ravishankar Shukla University, Raipur, 492010 (C.G.), India
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
- Research Institute for Biomedical Science, University of Hradec Králové, Antonína Dvoraka 451/1, 500 02 Hradec Kralove, Czech Republic
| |
Collapse
|
19
|
Pais MV, Kuo C, Ances BM, Wetherell JL, Lenze EJ, Diniz BS. Relationship between baseline plasma p-tau181 and longitudinal changes in cognition and structural brain measures in a cohort of cognitively unimpaired older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12487. [PMID: 37954547 PMCID: PMC10634375 DOI: 10.1002/dad2.12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/22/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Preclinical Alzheimer's disease (AD) affects a significant proportion of cognitively unimpaired (CU) older adults. Currently, blood-based biomarkers detect very early changes in the AD continuum with great accuracy. METHODS We measured baseline plasma phosphorylated tau (p-tau)181 using electrochemiluminescence (ECL)-based assay (MesoScale Discovery) in 533 CU older adults. Follow-up lasted up to 18 months. Cognitive performance assessment included memory and cognitive control. Structural brain measures included cortical thickness, which includes the AD magnetic resonance imaging (AD MRI) signature, and hippocampal volume. RESULTS In this cohort of CU older adults, baseline plasma p-tau181 levels were not associated with short-term changes in cognition and structural brain measures. Also, baseline plasma p-tau levels did not influence the effects of behavioral interventions (exercise or mindfulness) on cognitive and structural brain changes. DISCUSSION The short follow-up and healthy status of this CU cohort might have limited the sensitivity of plasma p-tau181 in detecting changes associated with AD pathology.
Collapse
Affiliation(s)
- Marcos V. Pais
- UConn Center on AgingUniversity of Connecticut Health CenterFarmingtonConnecticutUSA
- Laboratory of Neuroscience (LIM‐27)Departamento e Instituto de PsiquiatriaFaculdade de Medicina, Universidade de Sao Paulo (FMUSP)Sao PauloBrazil
| | - Chia‐Ling Kuo
- Department of Public Health SciencesUniversity of Connecticut Health CenterFarmingtonConnecticutUSA
| | - Beau M. Ances
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Eric J. Lenze
- Healthy Mind Lab, Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Breno S. Diniz
- UConn Center on AgingUniversity of Connecticut Health CenterFarmingtonConnecticutUSA
| |
Collapse
|
20
|
Mundada NS, Rojas JC, Vandevrede L, Thijssen EH, Iaccarino L, Okoye OC, Shankar R, Soleimani-Meigooni DN, Lago AL, Miller BL, Teunissen CE, Heuer H, Rosen HJ, Dage JL, Jagust WJ, Rabinovici GD, Boxer AL, La Joie R. Head-to-head comparison between plasma p-tau217 and flortaucipir-PET in amyloid-positive patients with cognitive impairment. Alzheimers Res Ther 2023; 15:157. [PMID: 37740209 PMCID: PMC10517500 DOI: 10.1186/s13195-023-01302-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Plasma phosphorylated tau (p-tau) has emerged as a promising biomarker for Alzheimer's disease (AD). Studies have reported strong associations between p-tau and tau-PET that are mainly driven by differences between amyloid-positive and amyloid-negative patients. However, the relationship between p-tau and tau-PET is less characterized within cognitively impaired patients with a biomarker-supported diagnosis of AD. We conducted a head-to-head comparison between plasma p-tau217 and tau-PET in patients at the clinical stage of AD and further assessed their relationships with demographic, clinical, and biomarker variables. METHODS We retrospectively included 87 amyloid-positive patients diagnosed with MCI or dementia due to AD who underwent structural MRI, amyloid-PET (11C-PIB), tau-PET (18F-flortaucipir, FTP), and blood draw assessments within 1 year (age = 66 ± 10, 48% female). Amyloid-PET was quantified in Centiloids (CL) while cortical tau-PET binding was measured using standardized uptake value ratios (SUVRs) referenced against inferior cerebellar cortex. Plasma p-tau217 concentrations were measured using an electrochemiluminescence-based assay on the Meso Scale Discovery platform. MRI-derived cortical volume was quantified with FreeSurfer. Mini-Mental State Examination (MMSE) scores were available at baseline (n = 85) and follow-up visits (n = 28; 1.5 ± 0.7 years). RESULTS Plasma p-tau217 and cortical FTP-SUVR were correlated (r = 0.61, p < .001), especially in temporo-parietal and dorsolateral frontal cortices. Both higher p-tau217 and FTP-SUVR values were associated with younger age, female sex, and lower cortical volume, but not with APOE-ε4 carriership. PIB-PET Centiloids were weakly correlated with FTP-SUVR (r = 0.26, p = 0.02), but not with p-tau217 (r = 0.10, p = 0.36). Regional PET-plasma associations varied with amyloid burden, with p-tau217 being more strongly associated with tau-PET in temporal cortex among patients with moderate amyloid-PET burden, and with tau-PET in primary cortices among patients with high amyloid-PET burden. Higher p-tau217 and FTP-SUVR values were independently associated with lower MMSE scores cross-sectionally, while only baseline FTP-SUVR predicted longitudinal MMSE decline when both biomarkers were included in the same model. CONCLUSION Plasma p-tau217 and tau-PET are strongly correlated in amyloid-PET-positive patients with MCI or dementia due to AD, and they exhibited comparable patterns of associations with demographic variables and with markers of downstream neurodegeneration.
Collapse
Affiliation(s)
- Nidhi S Mundada
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lawren Vandevrede
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Elisabeth H Thijssen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Ranjani Shankar
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Argentina L Lago
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Hillary Heuer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Howie J Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey L Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Global Brain Health Institute, San Francisco, CA, USA.
| |
Collapse
|