Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Blood biomarkers for Alzheimer’s disease in clinical practice and trials

Abstract

Blood-based biomarkers hold great promise to revolutionize the diagnostic and prognostic work-up of Alzheimer’s disease (AD) in clinical practice. This is very timely, considering the recent development of anti-amyloid-β (Aβ) immunotherapies. Several assays for measuring phosphorylated tau (p-tau) in plasma exhibit high diagnostic accuracy in distinguishing AD from all other neurodegenerative diseases in patients with cognitive impairment. Prognostic models based on plasma p-tau levels can also predict future development of AD dementia in patients with mild cognitive complaints. The use of such high-performing plasma p-tau assays in the clinical practice of specialist memory clinics would reduce the need for more costly investigations involving cerebrospinal fluid samples or positron emission tomography. Indeed, blood-based biomarkers already facilitate identification of individuals with pre-symptomatic AD in the context of clinical trials. Longitudinal measurements of such biomarkers will also improve the detection of relevant disease-modifying effects of new drugs or lifestyle interventions.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Suggested blood-based biomarker-based workflow for Alzheimer’s disease diagnostics.
Fig. 2: An overview of key blood-based biomarkers used in the diagnostic or prognostic work-up of Alzheimer’s disease.
Fig. 3: Suggested workflow for inclusion of study participants into preclinical Alzheimer’s disease trials.

Similar content being viewed by others

References

  1. DeTure, M. A. & Dickson, D. W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 14, 32 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hansson, O. Biomarkers for neurodegenerative diseases. Nat. Med. 27, 954–963 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Pichet Binette, A. et al. Amyloid-associated increases in soluble tau relate to tau aggregation rates and cognitive decline in early Alzheimer’s disease. Nat. Commun. 13, 6635 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Vogel, J. W. et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat. Med. 27, 871–881 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Budd Haeberlein, S. et al. Two randomized phase 3 studies of aducanumab in early Alzheimer’s disease. J. Prev. Alzheimers Dis. 9, 197–210 (2022).

    CAS  PubMed  Google Scholar 

  6. Mintun, M. A. et al. Donanemab in early Alzheimer’s disease. N. Engl. J. Med. 384, 1691–1704 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. van Dyck, C. H. et al. Lecanemab in early Alzheimer’s disease. N. Engl. J. Med. 388, 9–21 (2023).

    Article  PubMed  Google Scholar 

  8. Jansen, W. J. et al. Prevalence estimates of amyloid abnormality across the Alzheimer disease clinical spectrum. JAMA Neurol. 79, 228–243 (2022).

    Article  PubMed  Google Scholar 

  9. Smith, R., Wibom, M., Pawlik, D., Englund, E. & Hansson, O. Correlation of in vivo [18F]flortaucipir with postmortem Alzheimer disease tau pathology. JAMA Neurol. 76, 310–317 (2019).

    Article  PubMed  Google Scholar 

  10. Fleisher, A. S. et al. Positron emission tomography imaging with [18F]flortaucipir and postmortem assessment of Alzheimer disease neuropathologic changes. JAMA Neurol. 77, 829–839 (2020).

    Article  PubMed  Google Scholar 

  11. Ossenkoppele, R. et al. Discriminative accuracy of [18F]flortaucipir positron emission tomography for Alzheimer disease vs other neurodegenerative disorders. JAMA 320, 1151–1162 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ossenkoppele, R. & Hansson, O. Towards clinical application of tau PET tracers for diagnosing dementia due to Alzheimer’s disease. Alzheimers Dement. 17, 1998–2008 (2021).

    Article  PubMed  Google Scholar 

  13. Ossenkoppele, R. et al. Amyloid and tau PET-positive cognitively unimpaired individuals are at high risk for future cognitive decline. Nat. Med. 28, 2381–2387 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jack, C. R. Jr. et al. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 14, 535–562 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Janelidze, S. et al. CSF Aβ42/Aβ40 and Aβ42/Aβ38 ratios: better diagnostic markers of Alzheimer disease. Ann. Clin. Transl. Neurol. 3, 154–165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hansson, O. et al. CSF biomarkers of Alzheimer’s disease concord with amyloid-β PET and predict clinical progression: a study of fully automated immunoassays in BioFINDER and ADNI cohorts. Alzheimers Dement. 14, 1470–1481 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Mattsson-Carlgren, N. et al. Cerebrospinal fluid biomarkers in autopsy-confirmed Alzheimer disease and frontotemporal lobar degeneration. Neurology 98, e1137–e1150 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gobom, J. et al. Validation of the LUMIPULSE automated immunoassay for the measurement of core AD biomarkers in cerebrospinal fluid. Clin. Chem. Lab. Med. 60, 207–219 (2022).

    Article  CAS  PubMed  Google Scholar 

  19. Palmqvist, S. et al. Detailed comparison of amyloid PET and CSF biomarkers for identifying early Alzheimer disease. Neurology 85, 1240–1249 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Blennow, K. & Zetterberg, H. Biomarkers for Alzheimer’s disease: current status and prospects for the future. J. Intern. Med. 284, 643–663 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Janelidze, S. et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nat. Commun. 11, 1683 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Leuzy, A. et al. Comparing the clinical utility and diagnostic performance of CSF p-Tau181, p-Tau217, and p-Tau231 assays. Neurology 97, e1681–e1694 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Karikari, T. K. et al. Head-to-head comparison of clinical performance of CSF phospho-tau T181 and T217 biomarkers for Alzheimer’s disease diagnosis. Alzheimers Dement. 17, 755–767 (2021).

    Article  CAS  PubMed  Google Scholar 

  24. Suarez-Calvet, M. et al. 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. 12, e12921 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hanes, J. et al. Evaluation of a novel immunoassay to detect p-tau Thr217 in the CSF to distinguish Alzheimer disease from other dementias. Neurology 95, e3026–e3035 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mielke, M. M. et al. Comparison of CSF phosphorylated tau 181 and 217 for cognitive decline. Alzheimers Dement. 18, 602–611 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Mattsson-Carlgren, N. et al. The implications of different approaches to define AT(N) in Alzheimer disease. Neurology 94, e2233–e2244 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Schindler, S. E. et al. High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology 93, e1647–e1659 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Palmqvist, S., Mattsson, N., Hansson, O. & Alzheimer’s Disease Neuroimaging Initiative. Cerebrospinal fluid analysis detects cerebral amyloid-β accumulation earlier than positron emission tomography. Brain 139, 1226–1236 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Palmqvist, S. et al. Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in Alzheimer’s disease. EMBO Mol. Med. 11, e11170 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mattsson-Carlgren, N. 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. 6, eaaz2387 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Janelidze, S. et al. Associations of plasma phospho-tau217 levels with tau positron emission tomography in early Alzheimer disease. JAMA Neurol. 78, 149–156 (2021).

    Article  PubMed  Google Scholar 

  33. Groot, C. et al. Phospho-tau with subthreshold tau-PET predicts increased tau accumulation rates in amyloid-positive individuals. Brain https://doi.org/10.1093/brain/awac329 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Bastiaansen, A. E. M. et al. Autoimmune encephalitis resembling dementia syndromes. Neurol. Neuroimmunol. Neuroinflamm. 8, e1039 (2021).

  35. Riemenschneider, M. et al. Phospho-tau/total tau ratio in cerebrospinal fluid discriminates Creutzfeldt–Jakob disease from other dementias. Mol. Psychiatry 8, 343–347 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Hesse, C. et al. Transient increase in total tau but not phospho-tau in human cerebrospinal fluid after acute stroke. Neurosci. Lett. 297, 187–190 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Ost, M. et al. Initial CSF total tau correlates with 1-year outcome in patients with traumatic brain injury. Neurology 67, 1600–1604 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Khalil, M. et al. Neurofilaments as biomarkers in neurological disorders. Nat. Rev. Neurol. 14, 577–589 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Preische, O. et al. Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer’s disease. Nat. Med. 25, 277–283 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Salvado, G. et al. Optimal combinations of CSF biomarkers for predicting cognitive decline and clinical conversion in cognitively unimpaired participants and mild cognitive impairment patients: a multi-cohort study. Alzheimers Dement. https://doi.org/10.1002/alz.12907 (2023).

    Article  PubMed  Google Scholar 

  41. Salvadó, G. et al. Specific associations between plasma biomarkers and postmortem amyloid plaque and tau tangle loads. EMBO Mol. Med. https://doi.org/10.15252/emmm.202217123 (2023).

  42. Nakamura, A. et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature 554, 249–254 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Ovod, V. et al. Amyloid β concentrations and stable isotope labeling kinetics of human plasma specific to central nervous system amyloidosis. Alzheimers Dement. 13, 841–849 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Janelidze, S. et al. Plasma β-amyloid in Alzheimer’s disease and vascular disease. Sci. Rep. 6, 26801 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li, Y. et al. Validation of plasma amyloid-β 42/40 for detecting Alzheimer disease amyloid plaques. Neurology 98, e688–e699 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Palmqvist, S. et al. Performance of fully automated plasma assays as screening tests for Alzheimer disease-related β-amyloid status. JAMA Neurol. 76, 1060–1069 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  47. West, T. 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. 16, 30 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Janelidze, S. et al. Head-to-head comparison of 8 plasma amyloid-β 42/40 assays in Alzheimer disease. JAMA Neurol. 78, 1375–1382 (2021).

    Article  PubMed  Google Scholar 

  49. Roher, A. E. et al. Amyloid β peptides in human plasma and tissues and their significance for Alzheimer’s disease. Alzheimers Dement. 5, 18–29 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mielke, M. M. et al. Plasma phospho-tau181 increases with Alzheimer’s disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimers Dement. 14, 989–997 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Janelidze, 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. 26, 379–386 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Karikari, T. K. et al. Blood phosphorylated tau 181 as a biomarker for Alzheimer’s disease: a diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol. 19, 422–433 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Thijssen, E. H. et al. Diagnostic value of plasma phosphorylated tau181 in Alzheimer’s disease and frontotemporal lobar degeneration. Nat. Med. 26, 387–397 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Palmqvist, S. et al. Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 324, 772–781 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Barthelemy, N. R., Horie, K., Sato, C. & Bateman, R. J. Blood plasma phosphorylated-tau isoforms track CNS change in Alzheimer’s disease. J. Exp. Med. 217, e20200861 (2020).

  56. Ashton, N. J. et al. Plasma p-tau231: a new biomarker for incipient Alzheimer’s disease pathology. Acta Neuropathol. 141, 709–724 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Janelidze, S. et al. Head-to-head comparison of 10 plasma phospho-tau assays in prodromal Alzheimer’s disease. Brain https://doi.org/10.1093/brain/awac333 (2022).

    Article  PubMed Central  Google Scholar 

  58. Ashton, N. J. et al. Plasma and CSF biomarkers in a memory clinic: head-to-head comparison of phosphorylated tau immunoassays. Alzheimers Dement. https://doi.org/10.1002/alz.12841 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Mielke, M. M. et al. Comparison of plasma phosphorylated tau species with amyloid and tau positron emission tomography, neurodegeneration, vascular pathology, and cognitive outcomes. JAMA Neurol. 78, 1108–1117 (2021).

    Article  PubMed  Google Scholar 

  60. Bayoumy, S. et al. Clinical and analytical comparison of six Simoa assays for plasma p-tau isoforms p-tau181, p-tau217, and p-tau231. Alzheimers Res. Ther. 13, 198 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ashton, N. J. et al. Differential roles of Aβ42/40, p-tau231 and p-tau217 for Alzheimer’s trial selection and disease monitoring. Nat. Med. 28, 2555–2562 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mila-Aloma, M. et al. Plasma p-tau231 and p-tau217 as state markers of amyloid-β pathology in preclinical Alzheimer’s disease. Nat. Med. 28, 1797–1801 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Therriault, J. et al. Association of phosphorylated tau biomarkers with amyloid positron emission tomography vs tau positron emission tomography. JAMA Neurol. 80, 188–199 (2022).

    Article  PubMed Central  Google Scholar 

  64. Mattsson-Carlgren, N. et al. Soluble p-tau217 reflects amyloid and tau pathology and mediates the association of amyloid with tau. EMBO Mol. Med. 13, e14022 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Sato, C. et al. Tau kinetics in neurons and the human central nervous system. Neuron 98, 861–864 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kaeser, S. A. et al. CSF p-tau increase in response to Aβ-type and Danish-type cerebral amyloidosis and in the absence of neurofibrillary tangles. Acta Neuropathol. 143, 287–290 (2022).

    Article  CAS  PubMed  Google Scholar 

  67. Horie, K., Barthelemy, N. R., Sato, C. & Bateman, R. J. CSF tau microtubule binding region identifies tau tangle and clinical stages of Alzheimer’s disease. Brain 144, 515–527 (2021).

    Article  PubMed  Google Scholar 

  68. Ashton, N. J. et al. A multicentre validation study of the diagnostic value of plasma neurofilament light. Nat. Commun. 12, 3400 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gisslen, M. et al. Plasma concentration of the neurofilament light protein (NfL) is a biomarker of CNS injury in HIV infection: a cross-sectional study. EBioMedicine 3, 135–140 (2016).

    Article  PubMed  Google Scholar 

  70. Rodrigues, F. B. et al. Mutant huntingtin and neurofilament light have distinct longitudinal dynamics in Huntington’s disease. Sci. Transl. Med. 12, eabc2888 (2020).

  71. Verberk, I. M. W. et al. Combination of plasma amyloid β(1–42/1–40) and glial fibrillary acidic protein strongly associates with cerebral amyloid pathology. Alzheimers Res. Ther. 12, 118 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Pereira, J. B. et al. Plasma GFAP is an early marker of amyloid-β but not tau pathology in Alzheimer’s disease. Brain 144, 3505–3516 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Benedet, A. L. et al. Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer disease continuum. JAMA Neurol. 78, 1471–1483 (2021).

    Article  PubMed  Google Scholar 

  74. Verberk, I. M. W. 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. 2, e87–e95 (2021).

    Article  PubMed  Google Scholar 

  75. Cicognola, C. et al. Plasma glial fibrillary acidic protein detects Alzheimer pathology and predicts future conversion to Alzheimer dementia in patients with mild cognitive impairment. Alzheimers Res. Ther. 13, 68 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Heller, C. et al. Plasma glial fibrillary acidic protein is raised in progranulin-associated frontotemporal dementia. J. Neurol. Neurosurg. Psychiatry 91, 263–270 (2020).

    Article  PubMed  Google Scholar 

  77. Brum, W. S. et al. A two-step workflow based on plasma p-tau217 to screen for Aβ pathology with further confirmatory testing only in uncertain cases. Nat. Aging (in the press).

  78. Janelidze, S. et al. Detecting amyloid positivity in early Alzheimer’s disease using combinations of plasma Aβ42/Aβ40 and p-tau. Alzheimers Dement. 18, 283–293 (2022).

    Article  CAS  PubMed  Google Scholar 

  79. Palmqvist, S. et al. An accurate fully automated panel of plasma biomarkers for Alzheimer’s disease. Alzheimers Dement. https://doi.org/10.1002/alz.12751 (2022).

    Article  PubMed  Google Scholar 

  80. Karikari, T. K. et al. Blood phospho-tau in Alzheimer disease: analysis, interpretation, and clinical utility. Nat. Rev. Neurol. 18, 400–418 (2022).

    Article  CAS  PubMed  Google Scholar 

  81. Teunissen, C. E. et al. Blood-based biomarkers for Alzheimer’s disease: towards clinical implementation. Lancet Neurol. 21, 66–77 (2022).

    Article  CAS  PubMed  Google Scholar 

  82. Leuzy, A. et al. Blood-based biomarkers for Alzheimer’s disease. EMBO Mol. Med. 14, e14408 (2022).

    Article  CAS  PubMed  Google Scholar 

  83. Cullen, N. C. et al. Test–retest variability of plasma biomarkers in Alzheimer’s disease and its effects on clinical prediction models. Alzheimers Dement. 19, 797–806 (2023).

    Article  CAS  Google Scholar 

  84. Mielke, M. M. et al. Performance of plasma phosphorylated tau 181 and 217 in the community. Nat. Med. 28, 1398–1405 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Janelidze, S., Barthélemy, N. R., He, Y., Bateman, R. J. & Hansson, O. Mitigating the associations of kidney dysfunction with blood biomarkers of Alzheimer disease by using a phosphorylated tau to total tau ratio. JAMA Neurol. https://doi.org/10.1001/jamaneurol.2023.0199 (2023).

  86. Rabe, C. et al. Clinical performance and robustness evaluation of plasma amyloid-β42/40 prescreening. Alzheimers Dement. https://doi.org/10.1002/alz.12801 (2022).

    Article  PubMed  Google Scholar 

  87. Hansson, O. et al. The Alzheimer’s Association appropriate use recommendations for blood biomarkers in Alzheimer’s disease. Alzheimers Dement. 18, 2669–2686 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Mank, A. et al. Identifying relevant outcomes in the progression of Alzheimer’s disease; what do patients and care partners want to know about prognosis? Alzheimers Dement. 7, e12189 (2021).

    Google Scholar 

  89. Karikari, T. K. et al. Diagnostic performance and prediction of clinical progression of plasma phospho-tau181 in the Alzheimer’s Disease Neuroimaging Initiative. Mol. Psychiatry 26, 429–442 (2021).

    Article  CAS  PubMed  Google Scholar 

  90. Palmqvist, S. et al. Prediction of future Alzheimer’s disease dementia using plasma phospho-tau combined with other accessible measures. Nat. Med. 27, 1034–1042 (2021).

    Article  CAS  PubMed  Google Scholar 

  91. Cullen, N. et al. Individualized prognosis of cognitive decline and dementia in mild cognitive impairment based on plasma biomarker combinations. Nat. Aging 1, 114–123 (2021).

    Article  PubMed  Google Scholar 

  92. Smith, R. et al. Tau-PET is superior to phospho-tau when predicting cognitive decline in symptomatic AD patients. Alzheimers Dement. https://doi.org/10.1002/alz.12875 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Schindler, S. E. et al. Using Alzheimer’s disease blood tests to accelerate clinical trial enrollment. Alzheimers Dement. https://doi.org/10.1002/alz.12754 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Cullen, N. et al. Plasma amyloid-β42/40 and apolipoprotein E for amyloid PET pre-screening in secondary prevention trials of Alzheimer’s disease. Brain Commun. 5, fcad015 (2023).

  95. Insel, P. S. et al. Determining clinically meaningful decline in preclinical Alzheimer disease. Neurology 93, e322–e333 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  96. van der Kall, L. M. et al. Association of β-amyloid level, clinical progression, and longitudinal cognitive change in normal older individuals. Neurology 96, e662–e670 (2021).

    PubMed  PubMed Central  Google Scholar 

  97. Donohue, M. C. et al. Association between elevated brain amyloid and subsequent cognitive decline among cognitively normal persons. JAMA 317, 2305–2316 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Mattsson-Carlgren, N. et al. Prediction of longitudinal cognitive decline in preclinical Alzheimer disease using plasma biomarkers. JAMA Neurol. https://doi.org/10.1001/jamaneurol.2022.5272 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Ossenkoppele, R. et al. Accuracy of tau positron emission tomography as a prognostic marker in preclinical and prodromal Alzheimer disease: a head-to-head comparison against amyloid positron emission tomography and magnetic resonance imaging. JAMA Neurol. 78, 961–971 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Leuzy, A. et al. Biomarker-based prediction of longitudinal tau positron emission tomography in Alzheimer disease. JAMA Neurol. 79, 149–158 (2022).

    Article  PubMed  Google Scholar 

  101. Bradford, A., Kunik, M. E., Schulz, P., Williams, S. P. & Singh, H. Missed and delayed diagnosis of dementia in primary care: prevalence and contributing factors. Alzheimer Dis. Assoc. Disord. 23, 306–314 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  102. US Food and Drug Administration. Surrogate Endpoint Resources for Drug and Biologic Development https://www.fda.gov/drugs/development-resources/surrogate-endpoint-resources-drug-and-biologic-development (2018).

  103. Ning, L. & Wang, B. Neurofilament light chain in blood as a diagnostic and predictive biomarker for multiple sclerosis: a systematic review and meta-analysis. PLoS ONE 17, e0274565 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Zetterberg, H. et al. Association of cerebrospinal fluid neurofilament light concentration with Alzheimer disease progression. JAMA Neurol. 73, 60–67 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Mattsson, N., Andreasson, U., Zetterberg, H., Blennow, K. & Alzheimer’s Disease Neuroimaging, I. Association of plasma neurofilament light with neurodegeneration in patients with Alzheimer disease. JAMA Neurol. 74, 557–566 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Disanto, G. et al. Serum neurofilament light: a biomarker of neuronal damage in multiple sclerosis. Ann. Neurol. 81, 857–870 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Benatar, M. et al. Validation of serum neurofilaments as prognostic and potential pharmacodynamic biomarkers for ALS. Neurology 95, e59–e69 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Staffaroni, A. M. et al. Temporal order of clinical and biomarker changes in familial frontotemporal dementia. Nat. Med. 28, 2194–2206 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Delcoigne, B. et al. Blood neurofilament light levels segregate treatment effects in multiple sclerosis. Neurology 94, e1201–e1212 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Olsson, B. et al. NfL is a marker of treatment response in children with SMA treated with nusinersen. J. Neurol. 266, 2129–2136 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Salloway, S. et al. A trial of gantenerumab or solanezumab in dominantly inherited Alzheimer’s disease. Nat. Med. 27, 1187–1196 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Swanson, C. J. et al. A randomized, double-blind, phase 2b proof-of-concept clinical trial in early Alzheimer’s disease with lecanemab, an anti-Aβ protofibril antibody. Alzheimers Res. Ther. 13, 80 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Pontecorvo, M. J. et al. Association of donanemab treatment with exploratory plasma biomarkers in early symptomatic Alzheimer disease: a secondary analysis of the TRAILBLAZER-ALZ randomized clinical trial. JAMA Neurol. 79, 1250–1259 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Buchhave, P. 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 69, 98–106 (2012).

    Article  CAS  PubMed  Google Scholar 

  115. Samgard, K. et al. Cerebrospinal fluid total tau as a marker of Alzheimer’s disease intensity. Int. J. Geriatr. Psychiatry 25, 403–410 (2010).

    Article  PubMed  Google Scholar 

  116. Mattsson-Carlgren, N. et al. Longitudinal plasma p-tau217 is increased in early stages of Alzheimer’s disease. Brain 143, 3234–3241 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Hansson, O. et al. Plasma phosphorylated tau181 and neurodegeneration in Alzheimer’s disease. Ann. Clin. Transl. Neurol. 8, 259–265 (2021).

    Article  CAS  PubMed  Google Scholar 

  118. Moscoso, A. et al. Time course of phosphorylated-tau181 in blood across the Alzheimer’s disease spectrum. Brain 144, 325–339 (2021).

    Article  PubMed  Google Scholar 

  119. Portelius, E. et al. Ex vivo 18O-labeling mass spectrometry identifies a peripheral amyloid β clearance pathway. Mol. Neurodegener. 12, 18 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Yanamandra, K. et al. Anti-tau antibody administration increases plasma tau in transgenic mice and patients with tauopathy. Sci. Transl. Med. 9 eaal2029 (2017).

  121. Kuhlmann, J. et al. CSF Aβ1–42—an excellent but complicated Alzheimer’s biomarker—a route to standardisation. Clin. Chim. Acta 467, 27–33 (2017).

    Article  CAS  PubMed  Google Scholar 

  122. Boulo, S. et al. First amyloid β1–42 certified reference material for re-calibrating commercial immunoassays. Alzheimers Dement. 16, 1493–1503 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Pannee, J. et al. The global Alzheimer’s Association round robin study on plasma amyloid β methods. Alzheimers Dement. 13, e12242 (2021).

    Google Scholar 

  124. Andreasson, U. et al. Commutability of the certified reference materials for the standardization of β-amyloid 1–42 assay in human cerebrospinal fluid: lessons for tau and β-amyloid 1–40 measurements. Clin. Chem. Lab. Med. 56, 2058–2066 (2018).

    Article  CAS  PubMed  Google Scholar 

  125. Hansson, O. et al. The Alzheimer’s Association international guidelines for handling of cerebrospinal fluid for routine clinical measurements of amyloid β and tau. Alzheimers Dement. 17, 1575–1582 (2021).

    Article  CAS  PubMed  Google Scholar 

  126. Rozga, M., Bittner, T., Batrla, R. & Karl, J. Preanalytical sample handling recommendations for Alzheimer’s disease plasma biomarkers. Alzheimers Dement. 11, 291–300 (2019).

    Google Scholar 

  127. Verberk, I. M. W. et al. Characterization of pre-analytical sample handling effects on a panel of Alzheimer’s disease-related blood-based biomarkers: results from the Standardization of Alzheimer’s Blood Biomarkers (SABB) working group. Alzheimers Dement. 18, 1484–1497 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  128. Winston, C. N. et al. Evaluation of blood-based plasma biomarkers as potential markers of amyloid burden in preclinical Alzheimer’s disease. J. Alzheimers Dis. 92, 95–107 (2023).

    Article  CAS  PubMed  Google Scholar 

  129. Leuzy, A. et al. Robustness of CSF Aβ42/40 and Aβ42/p-tau181 measured using fully automated immunoassays to detect AD-related outcomes. Alzheimers Dement. https://doi.org/10.1002/alz.12897 (2023).

    Article  PubMed  Google Scholar 

  130. Benedet, A. L. et al. The accuracy and robustness of plasma biomarker models for amyloid PET positivity. Alzheimers Res. Ther. 14, 26 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Hampel, H. et al. Developing the ATX(N) classification for use across the Alzheimer disease continuum. Nat. Rev. Neurol. 17, 580–589 (2021).

    Article  PubMed  Google Scholar 

  132. Simren, J. et al. Establishment of reference values for plasma neurofilament light based on healthy individuals aged 5–90 years. Brain Commun. 4, fcac174 (2022).

    Google Scholar 

  133. Syrjanen, J. A. et al. Associations of amyloid and neurodegeneration plasma biomarkers with comorbidities. Alzheimers Dement. 18, 1128–1140 (2022).

    Article  CAS  PubMed  Google Scholar 

  134. Pichet Binette, A. et al. Confounding factors of Alzheimer’s disease plasma biomarkers and their impact on clinical performance. Alzheimers Dement. https://doi.org/10.1002/alz.12787 (2022).

    Article  PubMed  Google Scholar 

  135. Sullivan, G. M. & Feinn, R. Using effect size—or why the P value is not enough. J. Grad. Med. Educ. 4, 279–282 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  136. Lim, E., Miyamura, J. & Chen, J. J. Racial/ethnic-specific reference intervals for common laboratory tests: a comparison among Asians, Blacks, Hispanics, and white. Hawaii J. Med. Public Health 74, 302–310 (2015).

    PubMed  PubMed Central  Google Scholar 

  137. Brickman, A. M. et al. Plasma p-tau181, p-tau217, and other blood-based Alzheimer’s disease biomarkers in a multi-ethnic, community study. Alzheimers Dement. 17, 1353–1364 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Windon, C. et al. Comparison of plasma and CSF biomarkers across ethnoracial groups in the ADNI. Alzheimers Dement. 14, e12315 (2022).

    Google Scholar 

  139. Hu, Y. et al. Assessment of a plasma amyloid probability score to estimate amyloid positron emission tomography findings among adults with cognitive impairment. JAMA Netw. Open 5, e228392 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

O.H. was supported by the Swedish Research Council (2022-00775), ERA PerMed (ERAPERMED2021-184), the Knut and Alice Wallenberg Foundation (2017-0383), the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University, the Swedish Alzheimer Foundation (AF-980907), the Swedish Brain Foundation (FO2021-0293), the Parkinson Foundation of Sweden (1412/22), the Cure Alzheimer’s Fund, the Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, the Skåne University Hospital Foundation (2020-O000028), Regionalt Forskningsstöd (2022-1259) and the Swedish federal government under the ALF agreement (2022-Projekt0080). K.B. is supported by the Swedish Research Council (2017-00915 and 2022-00732; 1 January 2023 to 31 December 2026), the Alzheimer Drug Discovery Foundation, USA (RDAPB-201809-2016615), the Swedish Alzheimer Foundation (AF-930351, AF-939721 and AF-968270), Hjärnfonden, Sweden (FO2017-0243 and ALZ2022-0006), the Swedish state under the agreement between the Swedish government and the county councils, the ALF agreement (ALFGBG-715986 and ALFGBG-965240), the European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236), the National Institutes of Health, USA (grant 1R01AG068398-01), the Alzheimer’s Association 2021 Zenith Award (ZEN-21-848495) and the Alzheimer’s Association 2022–2025 grant (SG-23-1038904 QC). H.Z. is a Wallenberg Scholar supported by grants from the Swedish Research Council (2018-02532), the European Union’s Horizon Europe research and innovation program under grant agreement 101053962, Swedish State Support for Clinical Research (ALFGBG-71320), the Alzheimer Drug Discovery Foundation, USA (201809-2016862), the AD Strategic Fund and the Alzheimer’s Association (ADSF-21-831376-C, ADSF-21-831381-C and ADSF-21-831377-C), the Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (FO2022-0270), the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement 860197 (MIRIADE), the European Union Joint Programme, Neurodegenerative Disease Research (JPND2021-00694) and the UK Dementia Research Institute at UCL (UKDRI-1003).

Author information

Authors and Affiliations

Authors

Contributions

O.H. was mainly responsible for drafting and revising the manuscript, and he handled the correspondence. K.B., H.Z. and J.D. contributed substantially to the both drafting and revision of the manuscript.

Corresponding author

Correspondence to Oskar Hansson.

Ethics declarations

Competing interests

O.H. has acquired research support (for the institution) from ADx, Avid Radiopharmaceuticals, Biogen, Eli Lilly, Eisai, Fujirebio, GE Healthcare, Pfizer and Roche. In the past 2 years, he has received consultancy and/or speaker fees from AC Immune, Amylyx, ALZpath, BioArctic, Biogen, Cerveau, Eisai, Fujirebio, Genentech, Novartis, Roche and Siemens. K.B. has served as a consultant on advisory boards or on data-monitoring committees for Abcam, Axon, BioArctic, Biogen, JOMDD–Shimadzu, Julius Clinical, Lilly, MagQu, Novartis, Ono Pharma, Pharmatrophix, Prothena, Roche Diagnostics and Siemens Healthineers and is a cofounder of Brain Biomarker Solutions in Gothenburg (BBS), which is a part of the GU Ventures Incubator Program, outside the work presented in this paper. H.Z. has served on scientific advisory boards and/or as a consultant for AbbVie, Acumen, Alector, Alzinova, ALZpath, Annexon, Apellis, Artery Therapeutics, AZTherapies, CogRx, Denali, Eisai, NervGen, Novo Nordisk, Passage Bio, Pinteon Therapeutics, Prothena, Red Abbey Labs, reMYND, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics and Wave; has given lectures in symposia sponsored by Cellectricon, Fujirebio, AlzeCure, Biogen and Roche; and is a cofounder of Brain Biomarker Solutions in Gothenburg (BBS), which is a part of the GU Ventures Incubator Program (outside the work presented in this paper). J.D. is an inventor on patents or patent applications of Eli Lilly relating to the assays, methods, reagents and/or compositions of matter related to measurement of p-tau217. J.D. has served as a consultant for AbbVie, Genotix Biotechnologies, Gates Ventures, Karuna Therapeutics, ALZpath and Cognito Therapeutics and received research support from ADx NeuroSciences, Fujirebio, ALZpath, Roche Diagnostics and Eli Lilly in the past 2 years. J.D. has received speaker fees from Eli Lilly.

Peer review

Peer review information

Nature Aging thanks Elena Rodriguez-Vieitezand the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hansson, O., Blennow, K., Zetterberg, H. et al. Blood biomarkers for Alzheimer’s disease in clinical practice and trials. Nat Aging 3, 506–519 (2023). https://doi.org/10.1038/s43587-023-00403-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-023-00403-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing