Online first
Invited Review Article
Published online: 2024-08-02

open access

Page views 182
Article views/downloads 145
Get Citation

Connect on Social Media

Connect on Social Media

Clinical significance of plasma candidate biomarkers of Alzheimer’s Disease

Piotr Lewczuk12, Marta Łukaszewicz-Zając3, Johannes Kornhuber1, Barbara Mroczko23

Abstract

The number of patients with Alzheimer’s Disease (AD) has increased rapidly in recent decades. AD is a complex progressive neurodegenerative disease affecting c.14 million patients in Europe and the United States. The hallmarks of this disease are neurotic plaques composed of the amyloid-β (Aβ) peptide and neurofibrillary tangles formed of hyperphosphorylated tau protein (pTau). To date, four CSF biomarkers: amyloid beta 42 (Aβ42), Aβ42/40 ratio, Tau protein, and Tau phosphorylated at threonine 181 (pTau181) have been validated as core neurochemical AD biomarkers. Imaging biomarkers are valuable for AD diagnosis, although they suffer from limitations in their cost and accessibility, while CSF biomarkers require lumbar puncture. Thus, there is an urgent need for alternative, less invasive and more cost-effective biomarkers capable of diagnosing and monitoring AD progression in a clinical context, as well as expediting the development of new therapeutic strategies. This review assesses the potential clinical significance of plasma candidate biomarkers in AD diagnosis. We conclude that these proteins might hold great promise in identifying the pathological features of AD. However, the future implementation process, and validation of the assays’ accuracy using predefined cut-offs across more diverse patient populations, are crucial in establishing their utility in daily practice.

Article available in PDF format

View PDF Download PDF file

References

  1. World Health Organization, Dementia. https://www.who.int/news-room/fact-sheets/detail/dementia (18 Aug 2021).
  2. Feigin VL, Vos T, Nichols E, et al. The global burden of neurological disorders: translating evidence into policy. Lancet Neurol. 2020; 19(3): 255–265.
  3. Rocca WA, Petersen RC, Knopman DS, et al. Trends in the incidence and prevalence of Alzheimer's disease, dementia, and cognitive impairment in the United States. Alzheimers Dement. 2011; 7(1): 80–93.
  4. Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol. 2011; 7(3): 137–152.
  5. Dubois B, Feldman HH, Jacova C, et al. Research criteria for the diagnosis of Alzheimer's disease: revising the NINCDS-ADRDA criteria. Lancet Neurol. 2007; 6(8): 734–746.
  6. Petersen RC, Aisen PS, Beckett LA, et al. Alzheimer's Disease Neuroimaging Initiative (ADNI): clinical characterization. Neurology. 2010; 74(3): 201–209.
  7. Hansson O, Lehmann S, Otto M, et al. Advantages and disadvantages of the use of the CSF Amyloid β (Aβ) 42/40 ratio in the diagnosis of Alzheimer's Disease. Alzheimers Res Ther. 2019; 11(1): 34.
  8. Lewczuk P, Riederer P, O'Bryant SE, et al. Members of the WFSBP Task Force Working on this Topic: Peter Riederer, Carla Gallo, Dimitrios Kapogiannis, Andrea Lopez Mato, Florence Thibaut. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: An update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J Biol Psychiatry. 2018; 19(4): 244–328.
  9. Carroll CM, Li YM. Physiological and pathological roles of the γ-secretase complex. Brain Res Bull. 2016; 126(Pt 2): 199–206.
  10. Portelius E, Price E, Brinkmalm G, et al. A novel pathway for amyloid precursor protein processing. Neurobiol Aging. 2011; 32(6): 1090–1098.
  11. Lewczuk P, Matzen A, Blennow K, et al. Cerebrospinal Fluid Aβ42/40 Corresponds Better than Aβ42 to Amyloid PET in Alzheimer's Disease. J Alzheimers Dis. 2017; 55(2): 813–822.
  12. Olsson B, Lautner R, Andreasson U, et al. CSF and blood biomarkers for the diagnosis of Alzheimer's disease: a systematic review and meta-analysis. Lancet Neurol. 2016; 15(7): 673–684.
  13. Hulstaert F, Blennow K, Ivanoiu A, et al. Improved discrimination of AD patients using β-amyloid (1-42) and tau levels in CSF. Neurology. 1999; 52(8): 1555–1555.
  14. Galasko D, Chang L, Motter R, et al. High cerebrospinal fluid tau and low amyloid beta42 levels in the clinical diagnosis of Alzheimer disease and relation to apolipoprotein E genotype. Arch Neurol. 1998; 55(7): 937–945.
  15. Lewczuk P, Esselmann H, Bibl M, et al. Tau protein phosphorylated at threonine 181 in CSF as a neurochemical biomarker in Alzheimer's disease: original data and review of the literature. J Mol Neurosci. 2004; 23(1-2): 115–122.
  16. Blennow K, Zetterberg H. Biomarkers for Alzheimer's disease: current status and prospects for the future. J Intern Med. 2018; 284(6): 643–663.
  17. Lewczuk P, Esselmann H, Otto M, et al. Neurochemical diagnosis of Alzheimer's dementia by CSF Abeta42, Abeta42/Abeta40 ratio and total tau. Neurobiol Aging. 2004; 25(3): 273–281.
  18. Wiltfang J, Esselmann H, Bibl M, et al. Amyloid β peptide ratio 42/40 but not Aβ42 correlates with phospho‐Tau in patients with low‐ and high‐CSF Aβ40 load. Journal of Neurochemistry. 2006; 101(4): 1053–1059.
  19. Lewczuk P, Wiltfang J, Kornhuber J, et al. Distributions of Aβ42 and Aβ42/40 in the Cerebrospinal Fluid in View of the Probability Theory. Diagnostics (Basel). 2021; 11(12).
  20. Palmqvist S, Zetterberg H, Blennow K, et al. Accuracy of brain amyloid detection in clinical practice using cerebrospinal fluid β-amyloid 42: a cross-validation study against amyloid positron emission tomography. JAMA Neurol. 2014; 71(10): 1282–1289.
  21. Palmqvist S, Zetterberg H, Mattsson N, et al. Alzheimer's Disease Neuroimaging Initiative, Swedish BioFINDER Study Group. Detailed comparison of amyloid PET and CSF biomarkers for identifying early Alzheimer disease. Neurology. 2015; 85(14): 1240–1249.
  22. Palmqvist S, Mattsson N, Hansson O, et al. Alzheimer’s Disease Neuroimaging Initiative. Cerebrospinal fluid analysis detects cerebral amyloid-β accumulation earlier than positron emission tomography. Brain. 2016; 139(Pt 4): 1226–1236.
  23. Lewczuk P, Kornhuber J. Do we still need positron emission tomography for early Alzheimer's disease diagnosis? Brain. 2016; 139(11): e60.
  24. Henjum K, Almdahl IS, Årskog V, et al. Cerebrospinal fluid soluble TREM2 in aging and Alzheimer's disease. Alzheimers Res Ther. 2016; 8(1): 17.
  25. Mattsson N, Insel PS, Palmqvist S, et al. Increased amyloidogenic APP processing in APOE ɛ4-negative individuals with cerebral β-amyloidosis. Nat Commun. 2016; 7: 10918.
  26. Janelidze S, Zetterberg H, Mattsson N, et al. Swedish BioFINDER study group. CSF Aβ42/Aβ40 and Aβ42/Aβ38 ratios: better diagnostic markers of Alzheimer disease. Ann Clin Transl Neurol. 2016; 3(3): 154–165.
  27. Mulugeta E, Londos E, Ballard C, et al. CSF amyloid β38 as a novel diagnostic marker for dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2011; 82(2): 160–164.
  28. Lista S, Toschi N, Baldacci F, et al. Alzheimer Precision Medicine Initiative (APMI). Cerebrospinal Fluid Neurogranin as a Biomarker of Neurodegenerative Diseases: A Cross-Sectional Study. J Alzheimers Dis. 2017; 59(4): 1327–1334.
  29. Herskovits AZ, Locascio JJ, Peskind ER, et al. A Luminex assay detects amyloid β oligomers in Alzheimer's disease cerebrospinal fluid. PLoS One. 2013; 8(7): e67898.
  30. Fukumoto H, Tokuda T, Kasai T, et al. High-molecular-weight beta-amyloid oligomers are elevated in cerebrospinal fluid of Alzheimer patients. FASEB J. 2010; 24(8): 2716–2726.
  31. Gao CM, Yam AY, Wang X, et al. Aβ40 oligomers identified as a potential biomarker for the diagnosis of Alzheimer's disease. PLoS One. 2010; 5(12): e15725.
  32. Mandelkow E, von Bergen M, Biernat J, et al. Structural principles of tau and the paired helical filaments of Alzheimer's disease. Brain Pathol. 2007; 17(1): 83–90.
  33. Sunderland T, Linker G, Mirza N, et al. Decreased beta-amyloid1-42 and increased tau levels in cerebrospinal fluid of patients with Alzheimer disease. JAMA. 2003; 289(16): 2094–2103.
  34. Kohnken R, Buerger K, Zinkowski R, et al. Detection of tau phosphorylated at threonine 231 in cerebrospinal fluid of Alzheimer's disease patients. Neurosci Lett. 2000; 287(3): 187–190.
  35. Hampel H, Buerger K, Zinkowski R, et al. Measurement of phosphorylated tau epitopes in the differential diagnosis of Alzheimer disease: a comparative cerebrospinal fluid study. Arch Gen Psychiatry. 2004; 61(1): 95–102.
  36. Parnetti L, Lanari A, Amici S, et al. Phospho-Tau International Study Group. CSF phosphorylated tau is a possible marker for discriminating Alzheimer's disease from dementia with Lewy bodies. Phospho-Tau International Study Group. Neurol Sci. 2001; 22(1): 77–78.
  37. Arai H, Ishiguro K, Ohno H, et al. CSF phosphorylated tau protein and mild cognitive impairment: a prospective study. Exp Neurol. 2000; 166(1): 201–203.
  38. Suárez-Calvet M, Karikari TK, Ashton NJ, et al. ALFA Study. Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer's continuum when only subtle changes in Aβ pathology are detected. EMBO Mol Med. 2020; 12(12): e12921.
  39. Leuzy A, Janelidze S, Mattsson-Carlgren N, et al. Comparing the Clinical Utility and Diagnostic Performance of CSF P-Tau181, P-Tau217, and P-Tau231 Assays. Neurology. 2021; 97(17): e1681–e1694.
  40. Buchhave P, Minthon L, Zetterberg H, et al. Cerebrospinal fluid levels of β-amyloid 1-42, but not of tau, are fully changed already 5 to 10 years before the onset of Alzheimer dementia. Arch Gen Psychiatry. 2012; 69(1): 98–106.
  41. de Leon MJ, Pirraglia E, Osorio RS, et al. Alzheimer’s Disease Neuroimaging Initiative, National Alzheimer’s Coordinating Center. The nonlinear relationship between cerebrospinal fluid Aβ42 and tau in preclinical Alzheimer's disease. PLoS One. 2018; 13(2): e0191240.
  42. Molinuevo JL, Ayton S, Batrla R, et al. Current state of Alzheimer's fluid biomarkers. Acta Neuropathol. 2018; 136(6): 821–853.
  43. Shen Y, Wang H, Sun Q, et al. Increased Plasma Beta-Secretase 1 May Predict Conversion to Alzheimer's Disease Dementia in Individuals With Mild Cognitive Impairment. Biol Psychiatry. 2018; 83(5): 447–455.
  44. Leung YY, Toledo JB, Nefedov A, et al. Identifying amyloid pathology-related cerebrospinal fluid biomarkers for Alzheimer's disease in a multicohort study. Alzheimers Dement (Amst). 2015; 1(3): 339–348.
  45. Guo LH, Alexopoulos P, Perneczky R. Heart-type fatty acid binding protein and vascular endothelial growth factor: cerebrospinal fluid biomarker candidates for Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci. 2013; 263(7): 553–560.
  46. Heslegrave A, Heywood W, Paterson R, et al. Increased cerebrospinal fluid soluble TREM2 concentration in Alzheimer's disease. Mol Neurodegener. 2016; 11: 3.
  47. Baldacci F, Lista S, Cavedo E, et al. Diagnostic function of the neuroinflammatory biomarker YKL-40 in Alzheimer's disease and other neurodegenerative diseases. Expert Rev Proteomics. 2017; 14(4): 285–299.
  48. Muszyński P, Kulczyńska-Przybik A, Borawska R, et al. The Relationship between Markers of Inflammation and Degeneration in the Central Nervous System and the Blood-Brain Barrier Impairment in Alzheimer's Disease. J Alzheimers Dis. 2017; 59(3): 903–912.
  49. Iturria-Medina Y, Sotero RC, Toussaint PJ, et al. Alzheimer’s Disease Neuroimaging Initiative. Early role of vascular dysregulation on late-onsheimer’s disease based on multifactorial data-driven analysis. Nat Commun. 2016; 7: 11934.
  50. Pereira JB, Westman E, Hansson O. Alzheimer’s Disease Neuroimaging Initiative. Association between cerebrospinal fluid and plasma neurodegeneration biomarkers with brain atrophy in Alzheimer’s disease. Neurobiol Aging. 2017; 58: 14–29.
  51. Öhrfelt A, Brinkmalm A, Dumurgier J, et al. The pre-synaptic vesicle protein synaptotagmin is a novel biomarker for Alzheimer's disease. Alzheimers Res Ther. 2016; 8(1): 41.
  52. Chiasserini D, Biscetti L, Eusebi P, et al. Differential role of CSF fatty acid binding protein 3, α-synuclein, and Alzheimer's disease core biomarkers in Lewy body disorders and Alzheimer's dementia. Alzheimers Res Ther. 2017; 9(1): 52.
  53. Feneberg E, Steinacker P, Lehnert S, et al. Limited role of free TDP-43 as a diagnostic tool in neurodegenerative diseases. Amyotroph Lateral Scler Frontotemporal Degener. 2014; 15(5-6): 351–356.
  54. Mroczko B, Groblewska M, Zboch M, et al. Evaluation of visinin-like protein 1 concentrations in the cerebrospinal fluid of patients with mild cognitive impairment as a dynamic biomarker of Alzheimer's disease. J Alzheimers Dis. 2015; 43(3): 1031–1037.
  55. Zetterberg H, Skillbäck T, Mattsson N, et al. Alzheimer’s Disease Neuroimaging Initiative. Association of Cerebrospinal Fluid Neurofilament Light Concentration With Alzheimer Disease Progression. JAMA Neurol. 2016; 73(1): 60–67.
  56. Mroczko B, Groblewska M, Zboch M, et al. Concentrations of matrix metalloproteinases and their tissue inhibitors in the cerebrospinal fluid of patients with Alzheimer's disease. J Alzheimers Dis. 2014; 40(2): 351–357.
  57. Rohrer JD, Woollacott IOC, Dick KM, et al. Serum neurofilament light chain protein is a measure of disease intensity in frontotemporal dementia. Neurology. 2016; 87(13): 1329–1336.
  58. Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer's disease. Neuropeptides. 2015; 52: 1–18.
  59. Morris GP, Clark IA, Vissel B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer's disease. Acta Neuropathol Commun. 2014; 2: 135.
  60. Pedersen JT, Sigurdsson EM. Tau immunotherapy for Alzheimer's disease. Trends Mol Med. 2015; 21(6): 394–402.
  61. Lewczuk P, Lelental N, Lachmann I, et al. Non-Phosphorylated Tau as a Potential Biomarker of Alzheimer's Disease: Analytical and Diagnostic Characterization. J Alzheimers Dis. 2017; 55(1): 159–170.
  62. Ermann N, Lewczuk P, Schmitz M, et al. CSF nonphosphorylated Tau as a biomarker for the discrimination of AD from CJD. Ann Clin Transl Neurol. 2018; 5(7): 883–887.
  63. Foiani MS, Cicognola C, Ermann N, et al. Searching for novel cerebrospinal fluid biomarkers of tau pathology in frontotemporal dementia: an elusive quest. J Neurol Neurosurg Psychiatry. 2019; 90(7): 740–746.
  64. Parnetti L, Cicognola C, Eusebi P, et al. Value of cerebrospinal fluid α-synuclein species as biomarker in Parkinson's diagnosis and prognosis. Biomark Med. 2016; 10(1): 35–49.
  65. Eusebi P, Giannandrea D, Biscetti L, et al. Diagnostic utility of CSF α-synuclein species in Parkinson's disease: protocol for a systematic review and meta-analysis. BMJ Open. 2016; 6(6): e011113.
  66. Twohig D, Nielsen HM. α-synuclein in the pathophysiology of Alzheimer's disease. Mol Neurodegener. 2019; 14(1): 23.
  67. Slaets S, Vanmechelen E, Le Bastard N, et al. Increased CSF α-synuclein levels in Alzheimer's disease: correlation with tau levels. Alzheimers Dement. 2014; 10(5 Suppl): S290–S298.
  68. Engelborghs S, Niemantsverdriet E, Struyfs H, et al. Consensus guidelines for lumbar puncture in patients with neurological diseases. Alzheimers Dement (Amst). 2017; 8: 111–126.
  69. Hansson O, Mikulskis A, Fagan AM, et al. The impact of preanalytical variables on measuring cerebrospinal fluid biomarkers for Alzheimer's disease diagnosis: A review. Alzheimers Dement. 2018; 14(10): 1313–1333.
  70. Bjerke M, Andreasson U, Kuhlmann J, et al. Assessing the commutability of reference material formats for the harmonization of amyloid-β measurements. Clin Chem Lab Med. 2016; 54(7): 1177–1191.
  71. Lewczuk P, Zimmermann R, Wiltfang J, et al. Neurochemical dementia diagnostics: a simple algorithm for interpretation of the CSF biomarkers. J Neural Transm (Vienna). 2009; 116(9): 1163–1167.
  72. Lehmann S, Delaby C, Boursier G, et al. Relevance of Aβ42/40 Ratio for Detection of Alzheimer Disease Pathology in Clinical Routine: The PLM Scale. Front Aging Neurosci. 2018; 10: 138.
  73. Jack CR, Bennett DA, Blennow K, et al. A/T/N: An unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology. 2016; 87(5): 539–547.
  74. Lewczuk P, Kornhuber J, Toledo JB, et al. German Dementia Competence Network, US-ADNI. Validation of the Erlangen Score Algorithm for the Prediction of the Development of Dementia due to Alzheimer's Disease in Pre-Dementia Subjects. J Alzheimers Dis. 2015; 48(2): 433–441.
  75. Baldeiras I, Santana I, Leitão MJ, et al. Erlangen Score as a tool to predict progression from mild cognitive impairment to dementia in Alzheimer's disease. Alzheimers Res Ther. 2019; 11(1): 2.
  76. Skillbäck T, Kornhuber J, Blennow K, et al. Alzheimer’s Disease Neuroimaging Initiative. Erlangen Score Predicts Cognitive and Neuroimaging Progression in Mild Cognitive Impairment Stage of Alzheimer's Disease. J Alzheimers Dis. 2019; 69(2): 551–559.
  77. Somers C, Lewczuk P, Sieben A, et al. Validation of the Erlangen Score Algorithm for Differential Dementia Diagnosis in Autopsy-Confirmed Subjects. J Alzheimers Dis. 2019; 68(3): 1151–1159.
  78. Janelidze S, Berron D, Smith R, et al. Associations of Plasma Phospho-Tau217 Levels With Tau Positron Emission Tomography in Early Alzheimer Disease. JAMA Neurol. 2021; 78(2): 149–156.
  79. Beach TG, Monsell SE, Phillips LE, et al. Accuracy of the clinical diagnosis of Alzheimer disease at National Institute on Aging Alzheimer Disease Centers, 2005-2010. J Neuropathol Exp Neurol. 2012; 71(4): 266–273.
  80. Jack CR, Bennett DA, Blennow K, et al. Contributors. NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018; 14(4): 535–562.
  81. Palmqvist S, Stomrud E, Cullen N, et al. An accurate fully automated panel of plasma biomarkers for Alzheimer's disease. Alzheimers Dement. 2023; 19(4): 1204–1215.
  82. Dubois B, Villain N, Frisoni GB, et al. Clinical diagnosis of Alzheimer's disease: recommendations of the International Working Group. Lancet Neurol. 2021; 20(6): 484–496.
  83. Rabinovici GD, Carrillo MC, Apgar C, et al. Association of Amyloid Positron Emission Tomography With Subsequent Change in Clinical Management Among Medicare Beneficiaries With Mild Cognitive Impairment or Dementia. JAMA. 2019; 321(13): 1286–1294.
  84. Brand AL, Lawler PE, Bollinger JG, et al. The performance of plasma amyloid beta measurements in identifying amyloid plaques in Alzheimer's disease: a literature review. Alzheimers Res Ther. 2022; 14(1): 195.
  85. Bateman RJ, Munsell LY, Morris JC, et al. Human amyloid-beta synthesis and clearance rates as measured in cerebrospinal fluid in vivo. Nat Med. 2006; 12(7): 856–861.
  86. Hansson O. Biomarkers for neurodegenerative diseases. Nat Med. 2021; 27(6): 954–963.
  87. Ovod V, Ramsey KN, Mawuenyega KG, et al. Amyloid β concentrations and stable isotope labeling kinetics of human plasma specific to central nervous system amyloidosis. Alzheimers Dement. 2017; 13(8): 841–849.
  88. Teunissen CE, Verberk IMW, Thijssen EH, et al. Blood-based biomarkers for Alzheimer's disease: towards clinical implementation. Lancet Neurol. 2022; 21(1): 66–77.
  89. O'Bryant SE, Mielke MM, Rissman RA, et al. Biofluid Based Biomarker Professional Interest Area. Blood-based biomarkers in Alzheimer disease: Current state of the science and a novel collaborative paradigm for advancing from discovery to clinic. Alzheimers Dement. 2017; 13(1): 45–58.
  90. Pannee J, Törnqvist U, Westerlund A, et al. The amyloid-β degradation pattern in plasma--a possible tool for clinical trials in Alzheimer's disease. Neurosci Lett. 2014; 573: 7–12.
  91. Schindler SE, Bollinger JG, Ovod V, et al. High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology. 2019; 93(17): e1647–e1659.
  92. Pérez-Grijalba V, Arbizu J, Romero J, et al. AB255 Study Group. Plasma Aβ42/40 ratio alone or combined with FDG-PET can accurately predict amyloid-PET positivity: a cross-sectional analysis from the AB255 Study. Alzheimers Res Ther. 2019; 11(1): 96.
  93. Tanaka T, Ruifen JC, Nai YH, et al. Head-to-head comparison of amplified plasmonic exosome Aβ42 platform and single-molecule array immunoassay in a memory clinic cohort. Eur J Neurol. 2021; 28(5): 1479–1489.
  94. Smirnov DS, Ashton NJ, Blennow K, et al. Plasma biomarkers for Alzheimer's Disease in relation to neuropathology and cognitive change. Acta Neuropathol. 2022; 143(4): 487–503.
  95. Keshavan A, Pannee J, Karikari T, et al. Population-based blood screening for pre-clinical Alzheimer’s disease: a British birth cohort at age 70. Brain. 2021; 144(2): 434–449.
  96. Nakamura A, Kaneko N, Villemagne VL, et al. High performance plasma amyloid-β biomarkers for Alzheimer's disease. Nature. 2018; 554(7691): 249–254.
  97. West T, Kirmess KM, Meyer MR, et al. A blood-based diagnostic test incorporating plasma Aβ42/40 ratio, ApoE proteotype, and age accurately identifies brain amyloid status: findings from a multi cohort validity analysis. Mol Neurodegener. 2021; 16(1): 30.
  98. Janelidze S, Stomrud E, Palmqvist S, et al. Plasma β-amyloid in Alzheimer's disease and vascular disease. Sci Rep. 2016; 6: 26801.
  99. Hansson O, Zetterberg H, Vanmechelen E, et al. Evaluation of plasma Abeta(40) and Abeta(42) as predictors of conversion to Alzheimer’s disease in patients with mild cognitive impairment. Neurobiol Aging. 2010; 31: 357–67.
  100. Kuo YM, Emmerling MR, Lampert HC, et al. High levels of circulating Abeta42 are sequestered by plasma proteins in Alzheimer's disease. Biochem Biophys Res Commun. 1999; 257(3): 787–791.
  101. Mattsson N, Schöll M, Strandberg O, et al. 18F‐AV‐1451 and CSF T‐tau and P‐tau as biomarkers in Alzheimer's disease. EMBO Molecular Medicine. 2017; 9(9): 1212–1223.
  102. Mattsson-Carlgren N, Andersson E, Janelidze S, et al. Aβ deposition is associated with increases in soluble and phosphorylated tau that precede a positive Tau PET in Alzheimer's disease. Sci Adv. 2020; 6(16): eaaz2387.
  103. Meyer PF, Pichet Binette A, Gonneaud J, et al. Characterization of Alzheimer Disease Biomarker Discrepancies Using Cerebrospinal Fluid Phosphorylated Tau and AV1451 Positron Emission Tomography. JAMA Neurol. 2020; 77(4): 508–516.
  104. Bejanin A, Schonhaut DR, La Joie R, et al. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer's disease. Brain. 2017; 140(12): 3286–3300.
  105. Ossenkoppele R, Smith R, Ohlsson T, et al. Associations between tau, Aβ, and cortical thickness with cognition in Alzheimer disease. Neurology. 2019; 92(6).
  106. Mattsson N, Zetterberg H, Janelidze S, et al. ADNI Investigators. Plasma tau in Alzheimer disease. Neurology. 2016; 87(17): 1827–1835.
  107. Mielke MM, Hagen CE, Xu J, et al. Plasma phospho-tau181 increases with Alzheimer's disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimers Dement. 2018; 14(8): 989–997.
  108. Tatebe H, Kasai T, Ohmichi T, et al. Quantification of plasma phosphorylated tau to use as a biomarker for brain Alzheimer pathology: pilot case-control studies including patients with Alzheimer's disease and down syndrome. Mol Neurodegener. 2017; 12(1): 63.
  109. Yang CC, Chiu MJ, Chen TF, et al. Assay of Plasma Phosphorylated Tau Protein (Threonine 181) and Total Tau Protein in Early-Stage Alzheimer's Disease. J Alzheimers Dis. 2018; 61(4): 1323–1332.
  110. Coomans EM, Verberk IMW, Ossenkoppele R, et al. A Head-to-Head Comparison Between Plasma pTau181 and Tau PET Along the Alzheimer's Disease Continuum. J Nucl Med. 2023; 64(3): 437–443.
  111. Janelidze S, Mattsson N, Palmqvist S, et al. Plasma P-tau181 in Alzheimer's disease: relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer's dementia. Nat Med. 2020; 26(3): 379–386.
  112. Barthélemy NR, Bateman RJ, Hirtz C, et al. Cerebrospinal fluid phospho-tau T217 outperforms T181 as a biomarker for the differential diagnosis of Alzheimer's disease and PET amyloid-positive patient identification. Alzheimers Res Ther. 2020; 12(1): 26.
  113. Janelidze S, Stomrud E, Smith R, et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer's disease. Nat Commun. 2020; 11(1): 1683.
  114. Palmqvist S, Insel PS, Stomrud E, et al. Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in Alzheimer's disease. EMBO Mol Med. 2019; 11(12): e11170.
  115. Palmqvist S, Janelidze S, Quiroz YT, et al. Discriminative Accuracy of Plasma Phospho-tau217 for Alzheimer Disease vs Other Neurodegenerative Disorders. JAMA. 2020; 324(8): 772–781.
  116. Janelidze S, Bali D, Ashton NJ, et al. Head-to-head comparison of 10 plasma phospho-tau assays in prodromal Alzheimer's disease. Brain. 2023; 146(4): 1592–1601.
  117. Palmqvist S, Tideman P, Cullen N, et al. Alzheimer’s Disease Neuroimaging Initiative. Prediction of future Alzheimer's disease dementia using plasma phospho-tau combined with other accessible measures. Nat Med. 2021; 27(6): 1034–1042.
  118. Ashton NJ, Pascoal TA, Karikari TK, et al. Plasma p-tau231: a new biomarker for incipient Alzheimer's disease pathology. Acta Neuropathol. 2021; 141(5): 709–724.
  119. Brickman AM, Manly JJ, Honig LS, et al. Plasma p-tau181, p-tau217, and other blood-based Alzheimer's disease biomarkers in a multi-ethnic, community study. Alzheimers Dement. 2021; 17(8): 1353–1364.
  120. Karikari TK, Emeršič A, Vrillon A, et al. Head-to-head comparison of clinical performance of CSF phospho-tau T181 and T217 biomarkers for Alzheimer's disease diagnosis. Alzheimers Dement. 2021; 17(5): 755–767.
  121. Thijssen EH, La Joie R, Strom A, et al. Advancing Research and Treatment for Frontotemporal Lobar Degeneration investigators. Plasma phosphorylated tau 217 and phosphorylated tau 181 as biomarkers in Alzheimer's disease and frontotemporal lobar degeneration: a retrospective diagnostic performance study. Lancet Neurol. 2021; 20(9): 739–752.
  122. Chatterjee P, Pedrini S, Stoops E, et al. Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer’s disease. Translational Psychiatry. 2021; 11(1).
  123. Chatterjee P, Pedrini S, Doecke JD, et al. AIBL Research Group. Plasma Aβ42/40 ratio, p-tau181, GFAP, and NfL across the Alzheimer's disease continuum: A cross-sectional and longitudinal study in the AIBL cohort. Alzheimers Dement. 2023; 19(4): 1117–1134.
  124. Brum WS, Cullen NC, Therriault J, et al. A blood-based biomarker workflow for optimal tau-PET referral in memory clinic settings. Nat Commun. 2024; 15(1): 2311.
  125. Saitgareeva A, Bulygin K, Gareev I, et al. The role of microglia in the development of neurodegeneration. Neurological Sciences. 2020; 41(12): 3609–3615.
  126. Heneka M, Carson M, Khoury J, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol. 2015; 14(4): 388–405.
  127. Prins S, de Kam ML, Teunissen CE, et al. Inflammatory plasma biomarkers in subjects with preclinical Alzheimer's disease. Alzheimers Res Ther. 2022; 14(1): 106.
  128. Vergallo A, Lista S, Lemercier P, et al. INSIGHT-preAD study group and the Alzheimer Precision Medicine Initiative (APMI), INSIGHT-preAD study group, Alzheimer Precision Medicine Initiative (APMI). Association of plasma YKL-40 with brain amyloid-β levels, memory performance, and sex in subjective memory complainers. Neurobiol Aging. 2020; 96: 22–32.
  129. Kronborg G, Ostergaard C, Weis N, et al. Serum level of YKL-40 is elevated in patients with Streptococcus pneumoniae bacteremia and is associated with the outcome of the disease. Scand J Infect Dis. 2002; 34(5): 323–326.
  130. Verberk IMW, Laarhuis MB, van den Bosch KA, et al. Serum markers glial fibrillary acidic protein and neurofilament light for prognosis and monitoring in cognitively normal older people: a prospective memory clinic-based cohort study. Lancet Healthy Longev. 2021; 2(2): e87–e95.
  131. Prins S, Zhuparris A, Hart EP, et al. A cross-sectional study in healthy elderly subjects aimed at development of an algorithm to increase identification of Alzheimer pathology for the purpose of clinical trial participation. Alzheimers Res Ther. 2021; 13(1): 132.
  132. Pereira J, Janelidze S, Smith R, et al. Plasma GFAP is an early marker of amyloid-β but not tau pathology in Alzheimer’s disease. Brain. 2021; 144(11): 3505–3516.
  133. Bettcher BM, Fitch R, Wynn MJ, et al. MCP-1 and eotaxin-1 selectively and negatively associate with memory in MCI and Alzheimer's disease dementia phenotypes. Alzheimers Dement (Amst). 2016; 3: 91–97.
  134. Morgan AR, Touchard S, Leckey C, et al. NIMA Consortium, Annex: NIMA–Wellcome Trust Consortium for Neuroimmunology of Mood Disorders and Alzheimer's Disease. Inflammatory biomarkers in Alzheimer's disease plasma. Alzheimers Dement. 2019; 15(6): 776–787.
  135. Hu Q, Shi M, Li Y, et al. Elevated plasma neurofilament light was associated with multi-modal neuroimaging features in Alzheimer's Disease signature regions and predicted future tau deposition. Res Sq. 2024.
  136. Yuan A, Rao M, Nixon R. Neurofilaments and Neurofilament Proteins in Health and Disease. Cold Spring Harbor Perspectives in Biology. 2017; 9(4): a018309.
  137. Norgren N, Rosengren L, Stigbrand T. Elevated neurofilament levels in neurological diseases. Brain Research. 2003; 987(1): 25–31.
  138. Preische O, Schultz SA, Apel A, et al. Dominantly Inherited Alzheimer Network. Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer's disease. Nat Med. 2019; 25(2): 277–283.
  139. Mattsson N, Andreasson U, Zetterberg H, et al. Alzheimer’s Disease Neuroimaging Initiative. Association of Plasma Neurofilament Light With Neurodegeneration in Patients With Alzheimer Disease. JAMA Neurol. 2017; 74(5): 557–566.
  140. Mattsson N, Cullen NC, Andreasson U, et al. Association Between Longitudinal Plasma Neurofilament Light and Neurodegeneration in Patients With Alzheimer Disease. JAMA Neurol. 2019; 76(7): 791–799.
  141. Benedet A, Leuzy A, Pascoal T, et al. Stage-specific links between plasma neurofilament light and imaging biomarkers of Alzheimer’s disease. Brain. 2020; 143(12): 3793–3804.
  142. Nabizadeh F, Balabandian M, Rostami MR, et al. Alzheimer’s Disease Neuroimaging Initiative (ADNI). Plasma neurofilament light levels correlate with white matter damage prior to Alzheimer's disease: results from ADNI. Aging Clin Exp Res. 2022; 34(10): 2363–2372.
  143. Lewczuk P, Ermann N, Andreasson U, et al. Plasma neurofilament light as a potential biomarker of neurodegeneration in Alzheimer's disease. Alzheimers Res Ther. 2018; 10(1): 71.
  144. Clark C, Lewczuk P, Kornhuber J, et al. Plasma neurofilament light and phosphorylated tau 181 as biomarkers of Alzheimer's disease pathology and clinical disease progression. Alzheimers Res Ther. 2021; 13(1): 65.
  145. Chatterjee P, Pedrini S, Ashton NJ, et al. Diagnostic and prognostic plasma biomarkers for preclinical Alzheimer's disease. Alzheimers Dement. 2022; 18(6): 1141–1154.
  146. Gisslén M, Price RW, Andreasson U, et al. Plasma Concentration of the Neurofilament Light Protein (NFL) is a Biomarker of CNS Injury in HIV Infection: A Cross-Sectional Study. EBioMedicine. 2016; 3: 135–140.
  147. Weston PSJ, Poole T, Ryan NS, et al. Serum neurofilament light in familial Alzheimer disease: A marker of early neurodegeneration. Neurology. 2017; 89(21): 2167–2175.
  148. Blennow K, Zetterberg H. Understanding biomarkers of neurodegeneration: Ultrasensitive detection techniques pave the way for mechanistic understanding. Nat Med. 2015; 21(3): 217–219.
  149. Guo T, Shaw LM, Trojanowski JQ, et al. Alzheimer's Disease Neuroimaging Initiative. Association of CSF Aβ, amyloid PET, and cognition in cognitively unimpaired elderly adults. Neurology. 2020; 95(15): e2075–e2085.
  150. Luo J, Agboola F, Grant E, et al. Sequence of Alzheimer disease biomarker changes in cognitively normal adults: A cross-sectional study. Neurology. 2020; 95(23): e3104–e3116.