We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Theme: Alzheimer's disease: the biomarker revolution - Review

Cerebrospinal fluid biomarkers of Alzheimer’s disease

    Anne M Fagan

    † Author for correspondence

    Department of Neurology, Hope Center for Neurological Disorders and Alzheimer’s Disease Research Center, Washington University School of Medicine, 660 S. Euclid Ave., Box 8111, St Louis, MO 63110, USA.

    &
    Published Online:https://doi.org/10.2217/bmm.09.83

    Alzheimer’s disease will reach epidemic proportions within the next 20–30 years if left unchecked. Currently, there are no treatments that prevent or slow Alzheimer’s disease but many are being developed. Parallel efforts to develop biomarkers to aid in disease diagnosis and prognosis, and assess disease risk are currently underway. Clinicopathological and biomarker studies have demonstrated that Alzheimer’s disease pathology can be detected preclinically. Using biomarkers to identify affected individuals prior to the onset of clinical symptoms and associated synaptic/neuronal loss should enable novel clinical trial design and early mechanism-based therapeutic intervention. This article summarizes the most promising cerebrospinal fluid biomarkers, highlights novel applications and current challenges, and provides a prediction on how the field may evolve in 5–10 years.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Mirra S, Heyman A, McKeel D et al.: The consortium to establish a registry for Alzheimer’s disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology41,479–486 (1991).
    • Khachaturian Z: Diagnosis of Alzheimer’s disease. Arch. Neurol.42,1097–1105 (1985).
    • Hyman B, Trojanowski J: Consensus recommendations for the postmortem diagnosis of Alzheimer disease from the National Institute on Aging and the Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer disease. J. Neuropathol. Exp. Neurol.56,1095–1097 (1997).
    • Morris J: The clinical dementia rating (CDR). Current version and scoring rules. Neurology43,2412–2414 (1993).
    • Petersen R, Smith G, Waring S, Ivnik R, Tabgalos E, Kokmen E: Mild cognitive impairment: clinical characterization and outcome. Arch. Neurol.56,303–308 (1999).
    • Storandt M, Grant E, Miller J, Morris J: Longitudinal course and neuropathologic outcomes in original vs revised MCI and in pre-MCI. Neurology67,467–473 (2006).
    • Price J, Ko A, Wade M, Tsou S, McKeel D, Morris J: Neuron number in the entorhinal cortex and CA1 in preclinical Alzheimer’s disease. Arch. Neurol.58,1395–1402 (2001).
    • Morris J, Storandt M, Miller J et al.: Mild cognitive impairment represents early-stage Alzheimer’s disease. Arch. Neurol.58,397–405 (2001).
    • Cummings J, Jeste D: Alzheimer’s disease and its management in the year 2010. Psychiatr. Serv.50,1173–1177 (1999).
    • 10  Teel C: Rural practitioners’ experiences in dementia diagnosis and treatment. Aging Ment. Health8,422–429 (2004).
    • 11  Braak H, Braak E: Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol. Aging18,351–357 (1997).▪ A classic neuropathological study that defines the neuroanatomical development of Alzheimer’s disease (AD) neuropathological lesions with aging.
    • 12  Price J, Mckeel D, Buckles V et al.: Neuropathology of nondemented aging: Presumptive evidence for preclinical Alzheimer disease. Neurobiol. Aging30,1026–1036 (2009).
    • 13  Evans D, Funkenstein H, Albert M et al.: Prevalence of Alzheimer’s disease in a community population of older persons. Higher than previously reported. JAMA262,2551–2556 (1989).
    • 14  Morris J, Price J: Pathologic correlates of nondemented aging, mild cognitive impairment, and early stage Alzheimer’s disease. J. Mol. Neurosci.17,101–118 (2001).▪ Clinicopathological study correlating AD lesions with different stages of aging and development of AD dementia.
    • 15  Gomez-Isla T, Price J, McKeel D, Morris J, Growdon J, Hyman B: Profound loss of layer II entorhinal cortex neurons occurs in very mild Alzheimer’s disease. J. Neurosci.16,4491–4500 (1996).
    • 16  Markesbery W, Schmitt F, Kryscio R, Davis D, Smith C, Wekstein D: Neuropathologic substrate of mild cognitive impairment. Arch. Neurol.63,38–46 (2006).
    • 17  Brody D, Magnoni S, Schwetye K et al.: Amyloid-β dynamics correlate with neurological status in the injured human brain. Science321,1221–1224 (2008).▪ Elegant study using intracerebral microdialysis in living patients suffering from acute brain injury to measure analytes in serial samples of brain interstitial fluid and demonstrates a correlation between changes in amyloid-β (Aβ) concentration and neurological status.
    • 18  Hardy J, Selkoe D: The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science297,353–356 (2002).
    • 19  Wiltfang J, Esselmann H, Bibl M et al.: Amyloid β peptide ratio 42/40 but not Aβ 42 correlates with phospho-tau in patients with low- and high-CSF Aβ 40 load. J. Neurochem.101,1053–1059 (2007).
    • 20  Wiltfang J, Esselmann H, Bibl M et al.: Highly conserved and disease-specific patterns of carboxyterminally truncated Aβ peptides 1–37/38/39 in addition to 1–40/42 in Alzheimer’s disease and in patients with chronic neuroinflammation. J. Neurochem.81,481–496 (2002).
    • 21  Bibl M, Mollenhauer B, Esselmann H et al.: CSF amyloid-β-peptides in Alzheimer’s disease, dementia with Lewy bodies and Parkinson’s disease dementia. Brain129,1177–1187 (2006).
    • 22  Bibl M, Mollenhauer B, Esselmann H et al.: CSF diagnosis of Alzheimer’s disease and dementia with Lewy bodies. J. Neural Transm.10, (2006).
    • 23  Bibl M, Mollenhauer B, Lewczuk P et al.: Validation of amyloid-β peptides in CSF diagnosis of neurodegenerative dementias. Mol. Psychiatry12,671–680 (2007).
    • 24  Sunderland T, Linker G, Mirza N et al.: Decreased β-amyloid1–42 and increased tau levels in cerebrospinal fluid of patients with Alzheimer’s disease. JAMA289,2094–2103 (2003).
    • 25  Craig-Schapiro R, Fagan A, Holtzman D: Biomarkers of Alzheimer’s disease. Neurobiol. Dis.35,128–140 (2009).
    • 26  Mattsson N, Zetterberg H, Hansson O et al.: CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. JAMA302,385–393 (2009).
    • 27  Tapiola T, Alafuzoff I, Herukka S et al.: Cerebrospinal fluid β-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain. Arch. Neurol.66,382–389 (2009).
    • 28  Buerger K, Frisoni G, Uspenskaya O et al.: Validation of Alzheimer’s disease CSF and plasma biological markers: the multicentre reliability study of the pilot European Alzheimer’s Disease Neuroimaging Initiative (E-ADNI). Expl. Gerontol.44,579–585 (2009).
    • 29  Visser P, Verhey F, Knol D et al.: Prevalence and prognostic value of CSF markers of Alzheimer’s disease pathology in patients with subjective cognitive impairment or mild cognitive impairment in the DESCRIPA study: a prospective cohort study. Lancet Neurol.8,619–627 (2009).
    • 30  Blennow K, Hampel H: CSF markers for incipient Alzheimer’s disease. Lancet Neurol.2,605–613 (2003).
    • 31  Zhong Z, Ewers M, Teipel S et al.: Levels of b-secretase (BACE1) in cerebrospinal fluid as a predictor of risk in mild cognitive impairment. Arch. Gen. Psychiatry64,718–726 (2007).
    • 32  Ewers M, Zhong Z, Bürger K et al.: Increased CSF-BACE 1 activity is associated with APOE-ε4 genotype in subjects with mild cognitive impairment and Alzheimer’s disease. Brain131,1252–1258 (2008).
    • 33  Klunk W, Engler H, Nordberg A et al.: Imaging brain amyloid in Alzheimer’s disease with Pittsburgh compound-B. Ann. Neurol.55,306–319 (2004).▪▪Representing the first human study of amyloid-imaging PET tracer compound Pittsburgh Compound-B (PIB), this report describes retention of PIB in areas of AD brains known to contain amyloid, and additionally describes an inverse relationship between PIB PET signal and cerebral glucose metabolism, measured by FDG-PET.
    • 34  Fagan A, Mintun M, Mach R et al.: Inverse relation between in vivo amyloid imaging load and CSF Aβ42 in humans. Ann. Neurol.59,512–519 (2006).▪▪ Illustrates an inverse relationship between mean cortical retention of amyloid-imaging PET tracer compound PIB and cerebrospinal fluid (CSF) Aβ42 levels among demented and nondemented subjects alike, suggesting that brain amyloid deposition results in low CSF Aβ42, and that amyloid imaging and CSF Aβ42 might serve as antecedent biomarkers of preclinical AD.
    • 35  Fagan A, Roe C, Xiong C, Mintun M, Morris J, Holtzman D: Cerebrospinal fluid tau/Aβ42 ratio as a prediction of cognitive decline in nondemented older adults. Arch. Neurol.64,343–349 (2007).
    • 36  Forsberg A, Engler H, Almkvist O et al.: PET imaging of amyloid deposition in patients with mild cognitive impairment. Neurobiol. Aging29,1456–1465 (2008).
    • 37  Koivunen J, Pirttilä T, Kemppainen N et al.: PET amyloid ligand [11C]PIB uptake and cerebrospinal fluid β-amyloid in mild cognitive impairment. Dement. Geriatr. Cogn. Disord.26,378–383 (2008).
    • 38  Jagust W, Landau S, Shaw L et al.: Relationships between biomarkers in aging and dementia. Neurology73,1193–1199 (2009).
    • 39  Grimmer T, Riemenschneider M, Förstl H et al.: β amyloid in Alzheimer’s disease: increased deposition in brain is reflected in reduced concentration in cerebrospinal fluid. Biol. Psychiatry65,927–934 (2009).
    • 40  Fagan A, Mintun M, Shah A et al.: Cerebrospinal fluid tau and p-tau181 increase with cortical amyloid deposition in cognitively normal individuals: implications for future clinical trials of Alzheimer’s disease. EMBO Mol. Med.1,371–380 (2009).
    • 41  Aizenstein H, Nebes R, Saxton J et al.: Frequent amyloid deposition without significant cognitive impairment among the elderly. Arch. Neurol.65,1509–1517 (2008).
    • 42  Morris J, Roe C, Grant E et al.: Pittsburgh Compound B imaging and prediction of progression from cognitive normality to symptomatic Alzheimer disease. Arch. Neurol.66(12),1469–1475 (2009).▪▪ Demonstrates that the presence of brain amyloid in cognitively normal individuals is not benign but rather is predictive of future cognitive decline.
    • 43  Cairns N, Ikonomovic M, Benzinger T et al.: Absence of Pittsburgh Compound B detection of cerebral Amyloid β in a patient with clinical, cognitive, and cerebrospinal fluid markers of Alzheimer disease. Arch. Neurol.66(12),1557–1562 (2009).
    • 44  Itoh N, Arai H, Urakami K et al.: Large-scale, multicenter study of cerebrospinal fluid tau protein phosphorylated at serine 199 for the antemortem diagnosis of Alzheimer’s disease. Ann. Neurol.50,150–156 (2001).
    • 45  Buerger K, Teipel S, Zinkowski R et al.: CSF tau protein phosphorylated at threonine 231 correlates with cognitive decline in MCI subjects. Neurology59,627–629 (2002).
    • 46  Hampel H, Mitchell A, Blennow K et al.: Core biological marker candidates of Alzheimer’s disease – perspectives for diagnosis, prediction of outcome and reflection of biological activity. J. Neural. Transm.111,247–272 (2004).
    • 47  Buerger K, Ewers M, Pirttila T et al.: CSF phosphorylated tau protein correlates with neocortical neurofibrillary pathology in Alzheimer’s disease. Brain129(Pt 11),3035–3041 (2006).
    • 48  Ost M, Nylén K, Csajbok L et al.: Initial CSF total tau correlates with 1-year outcome in patients with traumatic brain injuries. Neurology67,1600–1604 (2006).
    • 49  Blennow K, Nellgård B: Amyloid β 1–42 and tau in cerebrospinal fluid after severe traumatic brain injury. Neurology62,159 (2004).
    • 50  Yao C, Williams A, Ottens A et al.: Detection of protein biomarkers using high-throughput immunoblotting following focal ischemic or penetrating ballistic-like brain injuries in rats. Brain Inj.22,723–732 (2008).
    • 51  Zetterberg H, Pedersen M, Lind K et al.: Intra-individual stability of CSF biomarkers for Alzheimer’s disease over two years. J. Alzheimers Dis.12,255–260 (2007).
    • 52  Blennow K, Zetterberg H, Minthon L et al.: Longitudinal stability of CSF biomarkers in Alzheimer’s disease. Neurosci. Lett.419,18–22 (2007).
    • 53  Terry RD, Masliah E, Salmon DP et al.: Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann. Neurol.30,572–580 (1991).
    • 54  Fagan A, Head D, Shah A et al.: Decreased cerebrospinal fluid Aβ42 correlates with brain atrophy in cognitively normal elderly. Ann. Neurol.65,175–183 (2009).
    • 55  Fukuyama R, Mizuno T, Mizuno T et al.: Age-dependent changes in the levels of Aβ40 and Aβ42 in cerebrospinal fluid from control subjects, and a decrease in the ratio of Aβ42 to Aβ40 level in cerebrospinal fluid from Alzheimer’s disease patients. Eur. Neurol.43,155–160 (2000).
    • 56  Mehta PD, Pirttila T, Mehta SP, Sersen EA, Aisen PS, Wisniewski HM: Plasma and cerebrospinal fluid levels of Amyloid β proteins 1–40 and 1–42 in Alzheimer’s disease. Arch. Neurol.57,100–105 (2000).
    • 57  Lewczuk P, Esselmann H, Otto M et al.: Neurochemical diagnosis of Alzheimer’s dementia by CSF Aβ42, Aβ42/Aβ40 ratio and total tau. Neurobiol. Aging25,273–281 (2004).
    • 58  Hansson O, Zetterberg H, Buchhave P et al.: Prediction of Alzheimer’s disease using the CSF Aβ42/Aβ40 ratio in patients with mild cognitive impairment. Dement. Geriatr. Cogn. Disord.23,316–320 (2007).
    • 59  Welge V, Fiege O, Lewczuk P et al.: Combined CSF tau, p-tau181 and amyloid-β 38/40/42 for diagnosing Alzheimer’s disease. J. Neural. Transm.116,203–212 (2009).
    • 60  Lewczuk P, Kornhuber J, Vanderstichele H et al.: Multiplexed quantification of dementia biomarkers in the CSF of patients with early dementias and MCI: A multicenter study. Neurobiol. Aging29,812–818 (2008).
    • 61  Hansson O, Zetterberg H, Buchhave P, Londos E, Blennow K, Minthon L: Association between CSF biomarkers and incipient Alzheimer’s disease in patients with mild cognitive impairment: a follow-up study. Lancet Neurol.5,228–234 (2006).▪ First report to demonstrate the utility of CSF biomarkers Aβ42, tau and phospho-tau to predict progression of patients from mild cognitive impairment to dementia attributed clinically to AD.
    • 62  Snider B, Fagan A, Roe C et al.: Cerebrospinal fluid biomarkers and rate of cognitive decline in very mild dementia of the Alzheimer type. Arch. Neurol.66,638–645 (2009).▪ First study to correlate CSF biomarkers with the rate of future cognitive decline in individuals diagnosed with very early stage dementia.
    • 63  Li G, Sokal I, Quinn J et al.: CSF tau/Aβ42 ratio for increased risk of mild cognitive impairment: a follow-up study. Neurology69,631–639 (2007).
    • 64  Zhang J, Goodlett D, Quinn J et al.: Quantitative proteomics of cerebrospinal fluid from patients with Alzheimer’s disease. J. Alzheimers Dis.7,125–133 (2005).
    • 65  Hu Y, Hosseini A, Kauwe J et al.: Identification and validation of novel CSF biomarkers for early stages of Alzheimer’s disease. Proteomics Clin. Appl.1,1373–1384 (2007).
    • 66  Castano E, Roher A, Esh C, Kokjohn T, Beach T: Comparative proteomics of cerebrospinal fluid in neuropathologically-confirmed Alzheimer’s disease and non-demented elderly subjects. Neurol. Res.28,155–163 (2006).
    • 67  Finehout E, Franck Z, Choe L, Relkin N, Lee K: Cerebrospinal fluid proteomic biomarkers for Alzheimer’s disease. Ann. Neurol.61,120–129 (2006).
    • 68  Archer H, Edison P, Brooks D et al.: Amyloid load and cerebral atrophy in Alzheimer’s disease: an 11C-PIB positron emission tomography study. Ann. Neurol.60,145–147 (2006).
    • 69  De Leon Mj, Desanti S, Zinkowski R et al.: Longitudinal CSF and MRI biomarkers improve the diagnosis of mild cognitive impairment. Neurobiol. Aging27(3),394–401 (2006).
    • 70  Hampel H, Burger K, Pruessner J et al.: Correlation of cerebrospinal fluid levels of tau protein phosphorylated at threonine 231 with rates of hippocampal atrophy in Alzheimer disease. Arch. Neurol.62,770–773 (2005).▪ Demonstrates that levels of CSF ptau231 can predict progression of hippocampal atrophy in AD, providing a good example of the power of combining biomarker modalities.
    • 71  De Leon M, Mosconi L, Li J et al.: Longitudinal CSF isoprostane and MRI atrophy in the progression to AD. J. Neurol. Sci.254(12),1666–1675 (2007).
    • 72  Kauwe J, Cruchaga C, Mayo K et al.: Variation in MAPT is associated with cerebrospinal fluid tau levels in the presence of amyloid-β deposition. Proc. Natl Acad. Sci. USA105,8050–8054 (2008).▪ Demonstrates that CSF biomarkers can be used as endophenotypes for genetic studies of AD by providing evidence for a genetic–epigenetic interaction that predisposes some individuals to the development of tauopathy and accelerated AD progression.
    • 73  Kauwe J, Wang J, Mayo K et al.: Alzheimer’s disease risk variants show association with cerebrospinal fluid amyloid β. Neurogenetics10,13–17 (2009).
    • 74  Bateman R, Munsell L, Morris J, Swarm R, Yarasheski K, Holtzman D: Human amyloid-β synthesis and clearance rates as measured in cerebrospinal fluid in vivo. Nat. Med.12,856–861 (2006).▪ This elegant study describes a novel method to measure the production and clearance rates of CNS proteins in living humans and demonstrated a very fast turnover of Aβ in young (23-45 years) cognitively normal individuals.
    • 75  Bateman R, Siemers E, Mawuenyega K et al.: A γ-secretase inhibitor decreases amyloid-β production in the central nervous system. Ann. Neurol.66,48–54 (2009).▪▪ This study demonstrates the utility of an in vivo labeling method to test the effect of a drug on the pharmacokinetics of CNS protein (in this case the Aβ peptide) production and clearance in living humans.
    • 76  Lewczuk P, Beck G, Esselmann H et al.: Effect of sample collection tubes on cerebrospinal fluid concentrations of tau proteins and amyloid β peptides. Clin. Chem.52,332–334 (2006).
    • 77  Peskind E, Nordberg A, Darreh-Shori T, Soininen H: Safety of lumbar puncture procedures in patients with Alzheimer’s disease. Curr. Alzheimer Res.6,290–292 (2009).
    • 78  Mueller S, Weiner M, Thal L et al.: Ways toward an early diagnosis in Alzheimer’s disease: the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Alzheimers Dement.1,55–66 (2005).▪ Introduces the Alzheimer’s Disease Neuroimaging Initiative (ADNI), the first large-scale, multi-institutional study of its kind designed to obtain longitudinal biomarker and clinical data from individuals with mild cognitive impairment, AD and cognitively normal individuals.
    • 79  Frisoni G, Henneman W, Weiner M et al.: The pilot European Alzheimer’s Disease Neuroimaging Initiative of the European Alzheimer’s Disease Consortium. Alzheimers Dement.4,255–264 (2008).
    • 80  Ihara Y: Overview on Alzheimer’s disease. Rinsho Shinkeigaku47,902–904 (2007).
    • 81  Coats M, Morris J: Antecedent biomarkers of Alzheimer’s disease: The Adult Children Study. J. Geriatr. Psychiatry Neurol.18,242–244 (2005).
    • 82  Kornhuber J, Schmidtke K, Frolich L et al.: Early and differential diagnosis of dementia and mild cognitive impairment: design and cohort baseline characteristics of the German Dementia Competence Network. Dement. Geriatr. Cogn. Disord.27,404–417 (2009).
    • 83  Perrin RJ, Fagan AM, Holtzman DM: Multimodal techniques for diagnosis and prognosis of Alzheimer’s disease. Nature461(7266),916–922 (2009).
    • 84  Fagan AM, Mintun MA, Shah AR et al.: Cerebrospinal fluid tau and p-tau181 increase with cortical amyloid deposition in cognitively normal individuals: implications for future clinical trials of Alzheimer’s disease. EMBO Mol. Med.1(8–9),371–380 (2009).
    • 85  Bateman RJ, Siemers ER, Mawuenyega KG et al.: A γ-secretase inhibitor decreases amyloid-β production in the central nervous system. Ann. Neurol.66,48–54 (2009).
    • 101  Alzheimer’s Association. www.alz.org
    • 102  Dominantly Inherited Alzheimer Network (DIAN). www.dian-info.org